151
|
Liang WH, Federico SM, London WB, Naranjo A, Irwin MS, Volchenboum SL, Cohn SL. Tailoring Therapy for Children With Neuroblastoma on the Basis of Risk Group Classification: Past, Present, and Future. JCO Clin Cancer Inform 2020; 4:895-905. [PMID: 33058692 PMCID: PMC7608590 DOI: 10.1200/cci.20.00074] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2020] [Indexed: 12/12/2022] Open
Abstract
For children with neuroblastoma, the likelihood of cure varies widely according to age at diagnosis, disease stage, and tumor biology. Treatments are tailored for children with this clinically heterogeneous malignancy on the basis of a combination of markers that are predictive of risk of relapse and death. Sequential risk-based, cooperative-group clinical trials conducted during the past 4 decades have led to improved outcome for children with neuroblastoma. Increasingly accurate risk classification and refinements in treatment stratification strategies have been achieved with the more recent discovery of robust genomic and molecular biomarkers. In this review, we discuss the history of neuroblastoma risk classification in North America and Europe and highlight efforts by the International Neuroblastoma Risk Group (INRG) Task Force to develop a consensus approach for pretreatment stratification using seven risk criteria including an image-based staging system-the INRG Staging System. We also update readers on the current Children's Oncology Group risk classifier and outline plans for the development of a revised 2021 Children's Oncology Group classifier that will incorporate INRG Staging System criteria to facilitate harmonization of risk-based frontline treatment strategies conducted around the globe. In addition, we discuss new approaches to establish increasingly robust, future risk classification algorithms that will further refine treatment stratification using machine learning tools and expanded data from electronic health records and the INRG Data Commons.
Collapse
Affiliation(s)
- Wayne H. Liang
- Department of Pediatrics and Informatics Institute, University of Alabama at Birmingham, Birmingham, AL
| | - Sara M. Federico
- Department of Oncology, St Jude Children’s Research Hospital, Memphis, TN
| | - Wendy B. London
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Arlene Naranjo
- Department of Biostatistics, Children’s Oncology Group Statistics and Data Center, University of Florida, Gainesville, FL
| | - Meredith S. Irwin
- Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Samuel L. Volchenboum
- Department of Pediatrics and Comer Children’s Hospital, University of Chicago, Chicago, IL
| | - Susan L. Cohn
- Department of Pediatrics and Comer Children’s Hospital, University of Chicago, Chicago, IL
| |
Collapse
|
152
|
Jin Z, Lu Y, Wu Y, Che J, Dong X. Development of differentiation modulators and targeted agents for treating neuroblastoma. Eur J Med Chem 2020; 207:112818. [PMID: 32937281 DOI: 10.1016/j.ejmech.2020.112818] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 08/10/2020] [Accepted: 09/03/2020] [Indexed: 02/07/2023]
Abstract
Neuroblastoma (NB) is one of the most common pediatric malignancies. Easy metastasis, poor prognosis, and a high degree of heterogeneity of NB hinder its successful treatment. Several different therapeutic strategies have been developed to overcome these problems, including differentiation and targeted therapy. In this review, we summarize the recent development of differentiation modulators and targeted agents for treating NB. Several promising targets of NB were also discussed.
Collapse
Affiliation(s)
- Zegao Jin
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Yang Lu
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Yizhe Wu
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Jinxin Che
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China.
| | - Xiaowu Dong
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China; Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310058, PR China; Cancer Center of Zhejiang University, Hangzhou, 310058, PR China.
| |
Collapse
|
153
|
Gains JE, Moroz V, Aldridge MD, Wan S, Wheatley K, Laidler J, Peet C, Bomanji JB, Gaze MN. A phase IIa trial of molecular radiotherapy with 177-lutetium DOTATATE in children with primary refractory or relapsed high-risk neuroblastoma. Eur J Nucl Med Mol Imaging 2020; 47:2348-2357. [PMID: 32157433 DOI: 10.1007/s00259-020-04741-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 02/20/2020] [Indexed: 12/15/2022]
Abstract
PURPOSE The objective of this phase IIa, open-label, single-centre, single-arm, two-stage clinical trial was to evaluate the safety and activity of 177-lutetium DOTATATE (LuDO) molecular radiotherapy in neuroblastoma. METHODS Children with relapsed or refractory metastatic high-risk neuroblastoma were treated with up to four courses of LuDO. The administered activity was 75 to 100 MBq kg-1 per course, spaced at 8- to 12-week intervals. Outcomes were assessed by the International Neuroblastoma Response Criteria (primary outcome), progression-free survival (PFS), and overall survival (OS). RESULTS The trial recruited 21 patients; eight received the planned four courses. There was dose-limiting haematologic toxicity in one case, but no other significant haematologic or renal toxicities. None of 14 evaluable patients had an objective response at 1 month after completion of treatment (Wilson 90% CI 0.0, 0.16; and 95% CI is 0.0, 0.22). The trial did not therefore proceed to the second stage. The median PFS was 2.96 months (95% CI 1.71, 7.66), and the median OS was 13.0 months (95% CI 2.99, 21.52). CONCLUSION In the absence of any objective responses, the use of LuDO as a single agent at the dose schedule used in this study is not recommended for the treatment of neuroblastoma. There are several reasons why this treatment schedule may not have resulted in objective responses, and as other studies do show benefit, the treatment should not be regarded as being of no value. Further trials designed to overcome this schedule's limitations are required. TRIAL REGISTRATION ISRCTN98918118; URL: https://www.isrctn.com/search?q=98918118.
Collapse
Affiliation(s)
- Jennifer E Gains
- Department of Oncology, University College London Hospitals NHS Foundation Trust, 250 Euston Road, London, NW1 2PG, UK
| | - Veronica Moroz
- Cancer Research UK Clinical Trials Unit, Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Matthew D Aldridge
- Department of Oncology, University College London Hospitals NHS Foundation Trust, 250 Euston Road, London, NW1 2PG, UK
- Department of Nuclear Medicine, University College London Hospitals NHS Foundation Trust, 235 Euston Road, London, NW1 2BU, UK
| | - Simon Wan
- Department of Nuclear Medicine, University College London Hospitals NHS Foundation Trust, 235 Euston Road, London, NW1 2BU, UK
| | - Keith Wheatley
- Cancer Research UK Clinical Trials Unit, Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Jennifer Laidler
- Cancer Research UK Clinical Trials Unit, Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Connie Peet
- Department of Oncology, University College London Hospitals NHS Foundation Trust, 250 Euston Road, London, NW1 2PG, UK
| | - Jamshed B Bomanji
- Department of Nuclear Medicine, University College London Hospitals NHS Foundation Trust, 235 Euston Road, London, NW1 2BU, UK
| | - Mark N Gaze
- Department of Oncology, University College London Hospitals NHS Foundation Trust, 250 Euston Road, London, NW1 2PG, UK.
| |
Collapse
|
154
|
Kawaguchi K, Umeda K, Takachi T, Ogura T, Horikoshi Y, Saida S, Kato I, Hiramatsu H, Adachi S, Takita J, Watanabe K. Effects of cryotherapy on high-dose melphalan-induced oral mucositis in pediatric patients undergoing autologous stem cell transplantation. Pediatr Blood Cancer 2020; 67:e28495. [PMID: 32573946 DOI: 10.1002/pbc.28495] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/26/2020] [Accepted: 05/27/2020] [Indexed: 01/20/2023]
Abstract
Cryotherapy is a conventional method for preventing melphalan-induced oral mucositis (OM) in adult patients. We retrospectively examined the clinical benefits of cryotherapy in 41 pediatric patients undergoing autologous stem cell transplantation using a melphalan-etoposide-carboplatin regimen. Twenty-two patients received cryotherapy. The cumulative incidence of grade 3-4 OM was significantly lower in the cryotherapy group (57.1%) than in the noncryotherapy group (89.5%; P = .041). Multivariate analyses identified cryotherapy and the melphalan dose as independent factors for the lower occurrence of OM. The present study demonstrates the clinically significant efficacy of cryotherapy for preventing melphalan-induced OM in pediatric patients.
Collapse
Affiliation(s)
- Koji Kawaguchi
- Department of Hematology and Oncology, Shizuoka Children's Hospital, Shizuoka, Japan
| | - Katsutsugu Umeda
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takayuki Takachi
- Department of Hematology and Oncology, Shizuoka Children's Hospital, Shizuoka, Japan
| | - Taemi Ogura
- Department of Hematology and Oncology, Shizuoka Children's Hospital, Shizuoka, Japan
| | - Yasuo Horikoshi
- Department of Hematology and Oncology, Shizuoka Children's Hospital, Shizuoka, Japan
| | - Satoshi Saida
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Itaru Kato
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hidefumi Hiramatsu
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Souichi Adachi
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Junko Takita
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kenichiro Watanabe
- Department of Hematology and Oncology, Shizuoka Children's Hospital, Shizuoka, Japan
| |
Collapse
|
155
|
Moreno L, Barone G, DuBois SG, Molenaar J, Fischer M, Schulte J, Eggert A, Schleiermacher G, Speleman F, Chesler L, Geoerger B, Hogarty MD, Irwin MS, Bird N, Blanchard GB, Buckland S, Caron H, Davis S, De Wilde B, Deubzer HE, Dolman E, Eilers M, George RE, George S, Jaroslav Š, Maris JM, Marshall L, Merchant M, Mortimer P, Owens C, Philpott A, Poon E, Shay JW, Tonelli R, Valteau-Couanet D, Vassal G, Park JR, Pearson ADJ. Accelerating drug development for neuroblastoma: Summary of the Second Neuroblastoma Drug Development Strategy forum from Innovative Therapies for Children with Cancer and International Society of Paediatric Oncology Europe Neuroblastoma. Eur J Cancer 2020; 136:52-68. [PMID: 32653773 DOI: 10.1016/j.ejca.2020.05.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 04/16/2020] [Accepted: 05/12/2020] [Indexed: 01/18/2023]
Abstract
Only one class of targeted agents (anti-GD2 antibodies) has been incorporated into front-line therapy for neuroblastoma since the 1980s. The Neuroblastoma New Drug Development Strategy (NDDS) initiative commenced in 2012 to accelerate the development of new drugs for neuroblastoma. Advances have occurred, with eight of nine high-priority targets being evaluated in paediatric trials including anaplastic lymphoma kinase inhibitors being investigated in front-line, but significant challenges remain. This article reports the conclusions of the second NDDS forum, which expanded across the Atlantic to further develop the initiative. Pre-clinical and clinical data for 40 genetic targets and mechanisms of action were prioritised and drugs were identified for early-phase trials. Strategies to develop drugs targeting TERT, telomere maintenance, ATRX, alternative lengthening of telomeres (ALT), BRIP1 and RRM2 as well as direct targeting of MYCN are high priority and should be championed for drug discovery. Promising pre-clinical data suggest that targeting of ALT by ATM or PARP inhibition may be potential strategies. Drugs targeting CDK2/9, CDK7, ATR and telomere maintenance should enter paediatric clinical development rapidly. Optimising the response to anti-GD2 by combinations with chemotherapy, targeted agents and other immunological targets are crucial. Delivering this strategy in the face of small patient cohorts, genomically defined subpopulations and a large number of permutations of combination trials, demands even greater international collaboration. In conclusion, the NDDS provides an internationally agreed, biologically driven selection of prioritised genetic targets and drugs. Improvements in the strategy for conducting trials in neuroblastoma will accelerate bringing these new drugs more rapidly to front-line therapy.
Collapse
Affiliation(s)
- Lucas Moreno
- Paediatric Haematology & Oncology Division, Hospital Universitari Vall d'Hebron, Barcelona, Spain.
| | - Giuseppe Barone
- Department of Paediatric Oncology, Great Ormond Street Hospital for Children, London, UK
| | - Steven G DuBois
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center and Harvard Medical School, Boston, MA, USA
| | - Jan Molenaar
- Princess Máxima Centre for Paediatric Oncology, Utrecht, The Netherlands
| | - Matthias Fischer
- Experimental Pediatric Oncology, University Children's Hospital, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Medical Faculty, University of Cologne, Cologne, Germany
| | - Johannes Schulte
- Department of Pediatric Oncology & Hematology, Charité University Hospital, Berlin, Germany
| | - Angelika Eggert
- Department of Pediatric Oncology & Hematology, Charité University Hospital, Berlin, Germany; German Cancer Consortium (DKTK Berlin), Berlin, Germany; Berlin Institute of Health (BIH), Berlin, Germany
| | - Gudrun Schleiermacher
- SIREDO, Department of Paediatric, Adolescents and Young Adults Oncology and INSERM U830, Institut Curie, Paris, France
| | - Frank Speleman
- Center for Medical Genetics Ghent (CMGG), Department of Biomolecular Medicine, Cancer Research Institute Ghent (CRIG), Belgium
| | - Louis Chesler
- Paediatric Drug Development, Children & Young People's Unit, The Royal Marsden NHS Foundation Trust, Sutton, UK; Division of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, Sutton, UK
| | - Birgit Geoerger
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Center, University Paris-Saclay & Inserm U1015, Villejuif, France
| | - Michael D Hogarty
- Division of Oncology, Children's Hospital of Philadelphia and Department of Pediatrics, University of Pennsylvania, USA; Perelman School of Medicine, University of Pennsylvania, USA
| | - Meredith S Irwin
- Department of Paediatrics, Medical Biophysics and Laboratory Medicine & Pathobiology, The Hospital for Sick Kids, Toronto, Canada
| | - Nick Bird
- Solving Kids' Cancer, UK and National Cancer Research Institute Children's Cancer & Leukaemia Clinical Studies Group, UK
| | - Guy B Blanchard
- Neuroblastoma UK & Department of Physiology, Development & Neuroscience, University of Cambridge, UK
| | | | | | | | - Bram De Wilde
- Center for Medical Genetics Ghent (CMGG), Department of Biomolecular Medicine, Cancer Research Institute Ghent (CRIG), Belgium
| | - Hedwig E Deubzer
- Center for Molecular Medicine Cologne (CMMC), Medical Faculty, University of Cologne, Cologne, Germany
| | - Emmy Dolman
- Department of Translational Research, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Martin Eilers
- Department of Biochemistry and Molecular Biology, University of Wuerzburg, Germany
| | - Rani E George
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center and Harvard Medical School, Boston, MA, USA
| | - Sally George
- Paediatric Drug Development, Children & Young People's Unit, The Royal Marsden NHS Foundation Trust, Sutton, UK; Division of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, Sutton, UK
| | - Štěrba Jaroslav
- Pediatric Oncology Department, University Hospital Brno, School of Medicine Masaryk University Brno, Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, ICRC Brno, St Anna University Hospital Brno, Czech Republic
| | - John M Maris
- Division of Oncology, Children's Hospital of Philadelphia and Department of Pediatrics, University of Pennsylvania, USA; Perelman School of Medicine, University of Pennsylvania, USA
| | - Lynley Marshall
- Paediatric Drug Development, Children & Young People's Unit, The Royal Marsden NHS Foundation Trust, Sutton, UK; Division of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, Sutton, UK
| | - Melinda Merchant
- Astrazeneca, Early Clinical Projects, Oncology Translation Medicines Unit, Innovative Medicines Unit, Cambridge, UK
| | - Peter Mortimer
- Astrazeneca, Early Clinical Projects, Oncology Translation Medicines Unit, Innovative Medicines Unit, Cambridge, UK
| | - Cormac Owens
- Department of Paediatric Haemaology/Oncology, Our Lady's Children's Hospital, Dublin, Ireland
| | | | - Evon Poon
- Division of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, Sutton, UK
| | - Jerry W Shay
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Roberto Tonelli
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Dominique Valteau-Couanet
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Center, University Paris-Saclay & Inserm U1015, Villejuif, France
| | - Gilles Vassal
- Department of Clinical Research, Gustave Roussy, Paris-Sud University, Paris, France
| | - Julie R Park
- Department of Pediatrics, University of Washington School of Medicine and Center for Clinical and Translational Research, Seattle Children's Hospital, USA
| | - Andrew D J Pearson
- Paediatric Drug Development, Children & Young People's Unit, The Royal Marsden NHS Foundation Trust, Sutton, UK; Division of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, Sutton, UK
| |
Collapse
|
156
|
Hishiki T, Fujino A, Watanabe T, Tahara K, Ohno M, Yamada Y, Tomonaga K, Kutsukake M, Fujita T, Kawakubo N, Matsumoto K, Kiyotani C, Shioda Y, Miyazaki O, Fuji H, Yoshioka T, Kanamori Y. Definitive Tumor Resection after Myeloablative High Dose Chemotherapy Is a Feasible and Effective Option in the Multimodal Treatment of High-Risk Neuroblastoma: A Single Institution Experience. J Pediatr Surg 2020; 55:1655-1659. [PMID: 31575417 DOI: 10.1016/j.jpedsurg.2019.08.050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 08/23/2019] [Accepted: 08/29/2019] [Indexed: 01/06/2023]
Abstract
BACKGROUND/PURPOSE The delayed local treatment approach (DL) in high-risk neuroblastoma (HR-NB) refers to the process in which tumor resection is performed after the completion of all the courses of chemotherapy, including myeloablative high-dose chemotherapy (HDC). Alternatively, in the conventional local treatment approach (CL), tumor resection is performed during induction chemotherapy. In this study, we compared the surgical outcomes in HR-NB patients treated by CL and DL. METHOD Forty-seven patients with abdominal HR-NB underwent primary tumor resection from 2002 to 2018. The timing of surgery was generally determined by following the trials and guidelines available at the time. The outcomes and surgical complications between the two strategies were compared. RESULT Operation time, blood loss, and postoperative WBC counts were lower in the DL group (n = 25) when compared to the CL group (n = 22), statistical significance notwithstanding. Major vascular structures were less frequently encased in the DL group tumors, while immediate surgical complications were significantly more frequent in the CL group (P < 0.05). Furthermore, the 3-year EFSs were 50.0% and 53.9% in the DL and CL groups, respectively. CONCLUSION DL appears to be a feasible and effective treatment option for HR-NB. Nonetheless, further verifications using larger cohorts are warranted. LEVEL OF EVIDENCE Treatment study, Level III.
Collapse
Affiliation(s)
- Tomoro Hishiki
- Division of Surgical Oncology, Children's Cancer Center, National Center for Child Health and Development; Division of Surgery, Department of Surgical Specialties, National Center for Child Health and Development; Department of Pediatric Surgical Oncology, National Cancer Center Hospital.
| | - Akihiro Fujino
- Division of Surgery, Department of Surgical Specialties, National Center for Child Health and Development
| | - Toshihiko Watanabe
- Division of Surgery, Department of Surgical Specialties, National Center for Child Health and Development
| | - Kazunori Tahara
- Division of Surgery, Department of Surgical Specialties, National Center for Child Health and Development
| | - Michinobu Ohno
- Division of Surgery, Department of Surgical Specialties, National Center for Child Health and Development
| | - Yohei Yamada
- Division of Surgery, Department of Surgical Specialties, National Center for Child Health and Development
| | - Kotaro Tomonaga
- Division of Surgery, Department of Surgical Specialties, National Center for Child Health and Development
| | - Mai Kutsukake
- Division of Surgery, Department of Surgical Specialties, National Center for Child Health and Development
| | - Takuro Fujita
- Division of Surgery, Department of Surgical Specialties, National Center for Child Health and Development
| | - Naonori Kawakubo
- Division of Surgical Oncology, Children's Cancer Center, National Center for Child Health and Development; Department of Pediatric Surgical Oncology, National Cancer Center Hospital
| | - Kimikazu Matsumoto
- Children's Cancer Center, National Center for Child Health and Development
| | - Chikako Kiyotani
- Children's Cancer Center, National Center for Child Health and Development
| | - Yoko Shioda
- Children's Cancer Center, National Center for Child Health and Development
| | - Osamu Miyazaki
- Department of Radiology, National Center for Child Health and Development
| | - Hiroshi Fuji
- Department of Radiology, National Center for Child Health and Development
| | - Takako Yoshioka
- Department of Pathology, National Center for Child Health and Development
| | - Yutaka Kanamori
- Division of Surgery, Department of Surgical Specialties, National Center for Child Health and Development
| |
Collapse
|
157
|
Serra-Roma A, Shakhova O. Identification of Novel Small-Molecule Kinase Modulators for the Treatment of Neuroblastoma. Oncol Ther 2020; 8:133-145. [PMID: 32700077 PMCID: PMC7359976 DOI: 10.1007/s40487-020-00113-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Indexed: 11/02/2022] Open
Abstract
Neuroblastoma represents 8-10% of all childhood cancer cases and is responsible for 15% of all cancer-related deaths in infants. Even though patients with low- and intermediate-risk disease have a good prognosis, the 5-year survival rate of the vast majority of patients with high-risk neuroblastoma is 50%. Despite extensive research efforts to find a cure for neuroblastoma, current treatment options are still limited. The aim of our study was to identify novel therapeutic compounds using high-throughput drug screening of a small molecule kinase inhibitor library containing 960 compounds. This screening resulted in the identification of two compounds, ST013381 and ST022328, that showed pronounced cytotoxic effects in six human neuroblastoma cell lines in vitro while having reduced effects in the BJ-5ta control cell line. These effects were observed in both MYCN-amplified and -non-amplified cells, indicating that these compounds can affect a wide range of neuroblastomas. Our experiments also revealed that several signaling pathways underlie the selective elimination of neuroblastoma cells by the ST013381 and ST022328 compounds. In summary, we have identified two novel compounds with a strong cytotoxic effect in vitro as promising agents for the treatment of neuroblastoma.
Collapse
Affiliation(s)
- André Serra-Roma
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Olga Shakhova
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
158
|
Ragoonanan D, Khazal SJ, Wang J, Payne A, Kohorst M, Harden A, Tewari P, Petropoulos D, Shoberu B, Kebriaei P, Mahadeo KM, Tambaro FP. Improved detection of sinusoidal obstructive syndrome using pediatric-AYA diagnostic criteria and severity grading. Bone Marrow Transplant 2020; 56:175-184. [PMID: 32665674 DOI: 10.1038/s41409-020-00998-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/23/2020] [Accepted: 07/02/2020] [Indexed: 01/19/2023]
Abstract
New diagnostic criteria and severity grading for sinusoidal obstructive syndrome (SOS) among pediatric and adolescent young adult (AYA) patients have been recently endorsed by international consensus. The extent to which these have been adopted in the US remains unclear. We sought to assess the potential impact via retrospective application of these criteria among patients treated at a large academic center in the United States. This is a single center retrospective study of pediatric-AYA patients who underwent hematopoietic cell transplantation (HCT) between July 2009 and 2019. The incidence of SOS was assessed using historic Baltimore and Seattle diagnostic criteria and compared with more recent guidelines (pEBMT) as proposed by the Paediatric Diseases Working Party of the European Society for Blood and Marrow Transplantation. Among 226 patients, application of the pEBMT diagnostic criteria was associated with a higher incidence (15.9%) and earlier time to diagnosis of SOS (by 2.5-3 days) compared with the modified Seattle (12.3%), and Baltimore (6.6%) criteria, respectively. The pEBMT criteria were sensitive and highly specific. Refractory thrombocytopenia was present in 75% of patients at diagnosis. Approximately 61% of patients with SOS were anicteric at diagnosis, though the majority (94.4%) developed hyperbilirubinemia as SOS progressed over a median time of 4 (1-57) days. Application of pEBMT criteria may have resulted in earlier indication for definitive treatment by 3 days. Timely diagnosis and administration of definitive treatment of SOS has been associated with improved outcomes. Prospective studies may better characterize the risk factors and natural course of SOS using pEBMT criteria.
Collapse
Affiliation(s)
- D Ragoonanan
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA.
| | - S J Khazal
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - J Wang
- Department of Biostatistics, Division of Basic Sciences, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - A Payne
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - M Kohorst
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - A Harden
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - P Tewari
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - D Petropoulos
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - B Shoberu
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - P Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas, 77030, USA
| | - K M Mahadeo
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - F P Tambaro
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA.,UOC SIT-TMO AORN Santobono-Pausilipon, Napoli, Italy
| |
Collapse
|
159
|
Holmes K, Pötschger U, Pearson ADJ, Sarnacki S, Cecchetto G, Gomez-Chacon J, Squire R, Freud E, Bysiek A, Matthyssens LE, Metzelder M, Monclair T, Stenman J, Rygl M, Rasmussen L, Joseph JM, Irtan S, Avanzini S, Godzinski J, Björnland K, Elliott M, Luksch R, Castel V, Ash S, Balwierz W, Laureys G, Ruud E, Papadakis V, Malis J, Owens C, Schroeder H, Beck-Popovic M, Trahair T, Forjaz de Lacerda A, Ambros PF, Gaze MN, McHugh K, Valteau-Couanet D, Ladenstein RL. Influence of Surgical Excision on the Survival of Patients With Stage 4 High-Risk Neuroblastoma: A Report From the HR-NBL1/SIOPEN Study. J Clin Oncol 2020; 38:2902-2915. [PMID: 32639845 DOI: 10.1200/jco.19.03117] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To evaluate the impact of surgeon-assessed extent of primary tumor resection on local progression and survival in patients in the International Society of Pediatric Oncology Europe Neuroblastoma Group High-Risk Neuroblastoma 1 trial. PATIENTS AND METHODS Patients recruited between 2002 and 2015 with stage 4 disease > 1 year or stage 4/4S with MYCN amplification < 1 year who had completed induction without progression, achieved response criteria for high-dose therapy (HDT), and had no resection before induction were included. Data were collected on the extent of primary tumor excision, severe operative complications, and outcome. RESULTS A total of 1,531 patients were included (median observation time, 6.1 years). Surgeon-assessed extent of resection included complete macroscopic excision (CME) in 1,172 patients (77%) and incomplete macroscopic resection (IME) in 359 (23%). Surgical mortality was 7 (0.46%) of 1,531. Severe operative complications occurred in 142 patients (9.7%), and nephrectomy was performed in 124 (8.8%). Five-year event-free survival (EFS) ± SE (0.40 ± 0.01) and overall survival (OS; 0.45 ± 0.02) were significantly higher with CME compared with IME (5-year EFS, 0.33 ± 0.03; 5-year OS, 0.37 ± 0.03; P < .001 and P = .004). The cumulative incidence of local progression (CILP) was significantly lower after CME (0.17 ± 0.01) compared with IME (0.30 ± 0.02; P < .001). With immunotherapy, outcomes were still superior with CME versus IME (5-year EFS, 0.47 ± 0.02 v 0.39 ± 0.04; P = .038); CILP was 0.14 ± 0.01 after CME and 0.27 ± 0.03 after IME (P < .002). A hazard ratio of 1.3 for EFS associated with IME compared with CME was observed before and after the introduction of immunotherapy (P = .030 and P = .038). CONCLUSION In patients with stage 4 high-risk neuroblastoma who have responded to induction therapy, CME of the primary tumor is associated with improved survival and local control after HDT, local radiotherapy (21 Gy), and immunotherapy.
Collapse
Affiliation(s)
- Keith Holmes
- Paediatric Surgery, St George's Hospital London and Royal Marsden Hospital, Sutton, United Kingdom
| | - Ulrike Pötschger
- Children's Cancer Research Institute, Department of Paediatrics, Medical University of Vienna, Vienna, Austria
| | - Andrew D J Pearson
- Institute of Cancer Research and Royal Marsden Hospital, Sutton, United Kingdom
| | - Sabine Sarnacki
- Department of Pediatric Surgery, Necker Enfants-Malades Hospital, Assistance Publique Hôpitaux de Paris, University de Paris, Paris, France
| | - Giovanni Cecchetto
- Pediatric Surgery, Department of Women's and Children's Health, University of Padua, Padua, Italy
| | - Javier Gomez-Chacon
- Paediatric Oncology, Paediatric Surgical Oncology Unit, Hospital Universitario La FE, Valencia, Spain
| | - Roly Squire
- Paediatric Oncology, Leeds Teaching Hospital, Leeds, United Kingdom
| | - Enrique Freud
- Schneider Children's Medical Center of Israel, Petach, Tikvah, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Adam Bysiek
- Department of Pediatric Surgery, University Children's Hospital, Kraków, Poland
| | - Lucas E Matthyssens
- Department of Gastrointestinal and Paediatric Surgery, Princess Elisabeth Children's Hospital, Ghent University Hospital, Ghent, Belgium
| | - Martin Metzelder
- Paediatric Surgery, Medical University of Vienna, Vienna, Austria
| | - Tom Monclair
- Oslo University Hospital Rikshospitalet, Oslo, Norway
| | | | - Michal Rygl
- University Hospital Motol, Prague, Czech Republic
| | - Lars Rasmussen
- Department of Surgical Gastroenterology A, Odense University Hospital, Odense, Denmark
| | | | - Sabine Irtan
- Sorbonne University, Department of Visceral and Neonatal Pediatric Surgery, Armand Trousseau Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Stefano Avanzini
- Pediatric Surgery Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Jan Godzinski
- Department of Paediatric Surgery, Marciniak Hospital, and Department of Paediatric Traumatology and Emergency Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Kristin Björnland
- Oslo University Hospital Rikshospitalet, Oslo, Norway.,University of Oslo, Oslo, Norway
| | - Martin Elliott
- Paediatric Oncology, Leeds Teaching Hospital, Leeds, United Kingdom
| | - Roberto Luksch
- Paediatric Oncology, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Victoria Castel
- Paediatric Oncology, Paediatric Surgical Oncology Unit, Hospital Universitario La FE, Valencia, Spain
| | - Shifra Ash
- Schneider Children's Medical Center of Israel, Petach, Tikvah, Israel
| | | | - Geneviève Laureys
- Department of Paediatric Haematology and Oncology, Princess Elisabeth Children's Hospital, Ghent University Hospital, Ghent, Belgium
| | - Ellen Ruud
- Oslo University Hospital Rikshospitalet, Oslo, Norway.,University of Oslo, Oslo, Norway
| | | | - Josef Malis
- University Hospital Motol, Prague, Czech Republic
| | - Cormac Owens
- Paediatric Haematology/Oncology, Our Lady's Children's Hospital, Crumlin, Dublin, Republic of Ireland
| | | | | | - Toby Trahair
- Sydney Children's Hospital, Randwick, New South Wales, Australia
| | | | - Peter F Ambros
- Children's Cancer Research Institute, Department of Paediatrics, Medical University of Vienna, Vienna, Austria
| | - Mark N Gaze
- University College Hospital, London, United Kingdom
| | - Kieran McHugh
- Paediatric Oncology, Great Ormond Street Hospital, London, United Kingdom
| | | | - Ruth Lydia Ladenstein
- St Anna Children's Hospital and Children's Cancer Research Institute, Department of Paediatrics, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
160
|
Wang J, Yao W, Li K. Applications and prospects of targeted therapy for neuroblastoma. WORLD JOURNAL OF PEDIATRIC SURGERY 2020; 3:e000164. [DOI: 10.1136/wjps-2020-000164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/07/2020] [Accepted: 05/21/2020] [Indexed: 11/04/2022] Open
Abstract
BackgroundNeuroblastoma is an extremely malignant tumor in children. For advanced or recurrent cases, existing treatment modalities are limited and efficacy remains disappointing. With the improvement in understanding of molecular biology of neuroblastoma and the development of clinical trials of targeted drug therapy, a variety of targeted therapies for neuroblastoma have appeared.Data sourcesAll the recent literatures on targeted therapies of neuroblastoma on PubMed were searched and reviewed.ResultsThis article reviewed targeted therapies of neuroblastoma going through clinical trials and obtained preliminary results. The features, advantages and disadvantages of targeted radiation therapy,immunotherapy, gene and pathway molecular inhibitor and angiogenesis inhibitor were discussed.ConclusionThis study provides references for better understanding the current progress of targeted therapies for neuroblastoma.
Collapse
|
161
|
Jain R, Hans R, Totadri S, Trehan A, Sharma RR, Menon P, Kapoor R, Saxena AK, Mittal BR, Bhatia P, Kakkar N, Srinivasan R, Rajwanshi A, Varma N, Samujh R, Marwaha N, Bansal D. Autologous stem cell transplant for high-risk neuroblastoma: Achieving cure with low-cost adaptations. Pediatr Blood Cancer 2020; 67:e28273. [PMID: 32196923 DOI: 10.1002/pbc.28273] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/16/2020] [Accepted: 03/02/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND The majority of patients in low- and middle-income countries (LMIC) are unable to receive optimal therapy, including autologous stem cell transplant (ASCT) for high-risk neuroblastoma. Management is intensive and multidisciplinary; survival is often poor. We report a single-center outcome of high-risk neuroblastoma, with adaptations optimized for LMIC. PROCEDURE The study was retrospective. Patients were treated on the backbone of the high-risk neuroblastoma study-1 of SIOP-Europe (HR-NBL1/SIOPEN) protocol with ASCT. Adaptations incorporated to decrease cost, requirement for inpatient admission, infections, and faster engraftment included (a) optional outpatient administration for rapid-COJEC, (b) two sessions of stem-cell apheresis, (c) storing stem cells at 2-6°C without cryopreservation for up to 7 days, (d) no central lines, (e) no antibacterial/antifungal/antiviral prophylaxis, (f) omitting formal assessment of cardiac/renal/pulmonary functions before ASCT, and (g) administration of pegylated granulocyte colony-stimulating factor on Day +4. RESULTS Over 5 years 9 months, 35 patients with high-risk neuroblastoma were treated. Rapid-COJEC was administered over a median duration of 80 days (interquartile range: 77, 83). Conditioning regimen included melphalan (n = 7), oral busulfan-melphalan (Bu/Mel; n = 6), or intravenous Bu/Mel (n = 22). The median viability of stem cells stored for 6 days (n = 28) was 93% (range: 88-99). Two (5.7%) patients had ASCT-related mortality. The 3-year overall and event-free survival was 41% and 39%, respectively. A relapse occurred in 20 (57%) patients. Treatment abandonment was observed in one (3%) patient. CONCLUSIONS Administration of therapy in a disciplined time frame along with low-cost adaptations enables to manage high-risk neuroblastoma with low abandonment and an encouraging survival in LMIC. Stem cells can be stored safely without cryopreservation for up to 7 days.
Collapse
Affiliation(s)
- Richa Jain
- Department of Pediatrics, Pediatric Hematology-Oncology Unit, Advanced Pediatrics Center, Chandigarh, India
| | - Rekha Hans
- Department of Transfusion, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Sidharth Totadri
- Department of Pediatrics, Pediatric Hematology-Oncology Unit, Advanced Pediatrics Center, Chandigarh, India
| | - Amita Trehan
- Department of Pediatrics, Pediatric Hematology-Oncology Unit, Advanced Pediatrics Center, Chandigarh, India
| | - Ratti Ram Sharma
- Department of Transfusion, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Prema Menon
- Department of Pediatric Surgery, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Rakesh Kapoor
- Department of Radiotherapy, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Akshay Kumar Saxena
- Departement of Radiodiagnosis, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Bhagwant Rai Mittal
- Department of Nuclear Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Prateek Bhatia
- Department of Pediatrics, Pediatric Hematology-Oncology Unit, Advanced Pediatrics Center, Chandigarh, India
| | - Nandita Kakkar
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Radhika Srinivasan
- Department of Cytopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Arvind Rajwanshi
- Department of Cytopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Neelam Varma
- Department of Hematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ram Samujh
- Department of Pediatric Surgery, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Neelam Marwaha
- Department of Transfusion, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Deepak Bansal
- Department of Pediatrics, Pediatric Hematology-Oncology Unit, Advanced Pediatrics Center, Chandigarh, India
| |
Collapse
|
162
|
George SL, Parmar V, Lorenzi F, Marshall LV, Jamin Y, Poon E, Angelini P, Chesler L. Novel therapeutic strategies targeting telomere maintenance mechanisms in high-risk neuroblastoma. J Exp Clin Cancer Res 2020; 39:78. [PMID: 32375866 PMCID: PMC7201617 DOI: 10.1186/s13046-020-01582-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 04/22/2020] [Indexed: 12/13/2022] Open
Abstract
The majority of high-risk neuroblastomas can be divided into three distinct molecular subgroups defined by the presence of MYCN amplification, upstream TERT rearrangements or alternative lengthening of telomeres (ALT). The common defining feature of all three subgroups is altered telomere maintenance; MYCN amplification and upstream TERT rearrangements drive high levels of telomerase expression whereas ALT is a telomerase independent telomere maintenance mechanism. As all three telomere maintenance mechanisms are independently associated with poor outcomes, the development of strategies to selectively target either telomerase expressing or ALT cells holds great promise as a therapeutic approach that is applicable to the majority of children with aggressive disease.Here we summarise the biology of telomere maintenance and the molecular drivers of aggressive neuroblastoma before describing the most promising therapeutic strategies to target both telomerase expressing and ALT cancers. For telomerase-expressing neuroblastoma the most promising targeted agent to date is 6-thio-2'-deoxyguanosine, however clinical development of this agent is required. In osteosarcoma cell lines with ALT, selective sensitivity to ATR inhibition has been reported. However, we present data showing that in fact ALT neuroblastoma cells are more resistant to the clinical ATR inhibitor AZD6738 compared to other neuroblastoma subtypes. More recently a number of additional candidate compounds have been shown to show selectivity for ALT cancers, such as Tetra-Pt (bpy), a compound targeting the telomeric G-quadruplex and pifithrin-α, a putative p53 inhibitor. Further pre-clinical evaluation of these compounds in neuroblastoma models is warranted.In summary, telomere maintenance targeting strategies offer a significant opportunity to develop effective new therapies, applicable to a large proportion of children with high-risk neuroblastoma. In parallel to clinical development, more pre-clinical research specifically for neuroblastoma is urgently needed, if we are to improve survival for this common poor outcome tumour of childhood.
Collapse
Affiliation(s)
- S L George
- Paediatric Tumour Biology, Division of Clinical Studies, The Institute of Cancer Research, London, UK.
- Children and Young People's Unit, Royal Marsden NHS Foundation Trust, London, UK.
| | - V Parmar
- Children and Young People's Unit, Royal Marsden NHS Foundation Trust, London, UK
| | - F Lorenzi
- Paediatric Tumour Biology, Division of Clinical Studies, The Institute of Cancer Research, London, UK
| | - L V Marshall
- Paediatric Tumour Biology, Division of Clinical Studies, The Institute of Cancer Research, London, UK
- Children and Young People's Unit, Royal Marsden NHS Foundation Trust, London, UK
| | - Y Jamin
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | - E Poon
- Paediatric Tumour Biology, Division of Clinical Studies, The Institute of Cancer Research, London, UK
| | - P Angelini
- Children and Young People's Unit, Royal Marsden NHS Foundation Trust, London, UK
| | - L Chesler
- Paediatric Tumour Biology, Division of Clinical Studies, The Institute of Cancer Research, London, UK
- Children and Young People's Unit, Royal Marsden NHS Foundation Trust, London, UK
| |
Collapse
|
163
|
Lau SCD, Unni MNM, Teh KH, Aziz MA, Muda Z, Thomas SA, Quah SW, Ibrahim H, Othman IS. Autologous stem cell transplantation following high-dose chemotherapy in children with high-risk neuroblastoma: Practicality in resource-limited countries. Pediatr Blood Cancer 2020; 67:e28176. [PMID: 31965709 DOI: 10.1002/pbc.28176] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/20/2019] [Accepted: 12/22/2019] [Indexed: 02/02/2023]
Affiliation(s)
- Sie Chong Doris Lau
- Department of Paediatrics, Hospital Wanita & Kanak-Kanak Kuala Lumpur, Kuala Lumpur, Malaysia.,Department of Paediatrics, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | | | - Kok Hoi Teh
- Department of Paediatrics, Hospital Wanita & Kanak-Kanak Kuala Lumpur, Kuala Lumpur, Malaysia
| | - Mimi Azura Aziz
- Department of Pathology, Hospital Wanita & Kanak-Kanak Kuala Lumpur, Kuala Lumpur, Malaysia
| | - Zulaiha Muda
- Department of Paediatrics, Hospital Wanita & Kanak-Kanak Kuala Lumpur, Kuala Lumpur, Malaysia
| | - Shoba Anne Thomas
- Department of Paediatrics, Hospital Wanita & Kanak-Kanak Kuala Lumpur, Kuala Lumpur, Malaysia
| | - Shiao Wei Quah
- Department of Paediatrics, Hospital Wanita & Kanak-Kanak Kuala Lumpur, Kuala Lumpur, Malaysia
| | - Hishamshah Ibrahim
- Department of Paediatrics, Hospital Wanita & Kanak-Kanak Kuala Lumpur, Kuala Lumpur, Malaysia
| | - Ida Shahnaz Othman
- Department of Paediatrics, Hospital Wanita & Kanak-Kanak Kuala Lumpur, Kuala Lumpur, Malaysia
| |
Collapse
|
164
|
Mody R, Yu AL, Naranjo A, Zhang FF, London WB, Shulkin BL, Parisi MT, Servaes SEN, Diccianni MB, Hank JA, Felder M, Birstler J, Sondel PM, Asgharzadeh S, Glade-Bender J, Katzenstein H, Maris JM, Park JR, Bagatell R. Irinotecan, Temozolomide, and Dinutuximab With GM-CSF in Children With Refractory or Relapsed Neuroblastoma: A Report From the Children's Oncology Group. J Clin Oncol 2020; 38:2160-2169. [PMID: 32343642 DOI: 10.1200/jco.20.00203] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PURPOSE The combination of irinotecan, temozolomide, dintuximab, and granulocyte-macrophage colony-stimulating factor (I/T/DIN/GM-CSF) demonstrated activity in patients with relapsed/refractory neuroblastoma in the randomized Children's Oncology Group ANBL1221 trial. To more accurately assess response rate and toxicity, an expanded cohort was nonrandomly assigned to I/T/DIN/GM-CSF. PATIENTS AND METHODS Patients were eligible at first relapse or first designation of refractory disease. Oral T and intravenous (IV) irinotecan were administered on days 1 to 5 of 21-day cycles. DIN was administered IV (days 2-5), and GM-CSF was administered subcutaneously (days 6-12). The primary end point was objective response, analyzed on an intent-to-treat basis per the International Neuroblastoma Response Criteria. RESULTS Seventeen eligible patients were randomly assigned to I/T/DIN/GM-CSF (February 2013 to March 2015); 36 additional patients were nonrandomly assigned to I/T/DIN/GM-CSF (August 2016 to May 2017). Objective (complete or partial) responses were observed in nine (52.9%) of 17 randomly assigned patients (95% CI, 29.2% to 76.7%) and 13 (36.1%) of 36 expansion patients (95% CI, 20.4% to 51.8%). Objective responses were seen in 22 (41.5%) of 53 patients overall (95% CI, 28.2% to 54.8%); stable disease was also observed in 22 of 53. One-year progression-free and overall survival for all patients receiving I/T/DIN/GM-CSF were 67.9% ± 6.4% (95% CI, 55.4% to 80.5%) and 84.9% ± 4.9% (95% CI, 75.3% to 94.6%), respectively. Two patients did not receive protocol therapy and were evaluable for response but not toxicity. Common grade ≥ 3 toxicities were fever/infection (18 [35.3%] of 51), neutropenia (17 [33.3%] of 51), pain (15 [29.4%] of 51), and diarrhea (10 [19.6%] of 51). One patient met protocol-defined criteria for unacceptable toxicity (grade 4 hypoxia). Higher DIN trough levels were associated with response. CONCLUSION I/T/DIN/GM-CSF has significant antitumor activity in patients with relapsed/refractory neuroblastoma. Study of chemoimmunotherapy in the frontline setting is indicated, as is further evaluation of predictive biomarkers.
Collapse
Affiliation(s)
- Rajen Mody
- C.S. Mott Children's Hospital, University of Michigan, Ann Arbor, MI
| | - Alice L Yu
- University of California San Diego, San Diego, CA.,Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou and Chang Gung University, Taiwan
| | - Arlene Naranjo
- Children's Oncology Group Statistics and Data Center, University of Florida, Gainesville, FL
| | - Fan F Zhang
- Children's Oncology Group Statistics and Data Center, Monrovia, CA
| | - Wendy B London
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Barry L Shulkin
- St Jude Children's Research Hospital and University of Tennessee Health Science Center, Memphis, TN
| | | | - Sabah-E-Noor Servaes
- Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| | | | | | | | | | | | - Shahab Asgharzadeh
- Children's Hospital of Los Angeles and University of Southern California, Los Angeles, CA
| | | | | | - John M Maris
- Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| | - Julie R Park
- Seattle Children's Hospital and University of Washington, Seattle, WA
| | - Rochelle Bagatell
- Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
165
|
Martí-Bonmatí L, Alberich-Bayarri Á, Ladenstein R, Blanquer I, Segrelles JD, Cerdá-Alberich L, Gkontra P, Hero B, García-Aznar JM, Keim D, Jentner W, Seymour K, Jiménez-Pastor A, González-Valverde I, Martínez de Las Heras B, Essiaf S, Walker D, Rochette M, Bubak M, Mestres J, Viceconti M, Martí-Besa G, Cañete A, Richmond P, Wertheim KY, Gubala T, Kasztelnik M, Meizner J, Nowakowski P, Gilpérez S, Suárez A, Aznar M, Restante G, Neri E. PRIMAGE project: predictive in silico multiscale analytics to support childhood cancer personalised evaluation empowered by imaging biomarkers. Eur Radiol Exp 2020; 4:22. [PMID: 32246291 PMCID: PMC7125275 DOI: 10.1186/s41747-020-00150-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 02/24/2020] [Indexed: 03/12/2023] Open
Abstract
PRIMAGE is one of the largest and more ambitious research projects dealing with medical imaging, artificial intelligence and cancer treatment in children. It is a 4-year European Commission-financed project that has 16 European partners in the consortium, including the European Society for Paediatric Oncology, two imaging biobanks, and three prominent European paediatric oncology units. The project is constructed as an observational in silico study involving high-quality anonymised datasets (imaging, clinical, molecular, and genetics) for the training and validation of machine learning and multiscale algorithms. The open cloud-based platform will offer precise clinical assistance for phenotyping (diagnosis), treatment allocation (prediction), and patient endpoints (prognosis), based on the use of imaging biomarkers, tumour growth simulation, advanced visualisation of confidence scores, and machine-learning approaches. The decision support prototype will be constructed and validated on two paediatric cancers: neuroblastoma and diffuse intrinsic pontine glioma. External validation will be performed on data recruited from independent collaborative centres. Final results will be available for the scientific community at the end of the project, and ready for translation to other malignant solid tumours.
Collapse
Affiliation(s)
- Luis Martí-Bonmatí
- Medical Imaging Department, La Fe University and Polytechnic Hospital & Biomedical Imaging Research Group (GIBI230) at La Fe University and Polytechnic Hospital and Health Research Institute, Av. Fernando Abril Martorell 106, 46026, Valencia, Spain.
| | - Ángel Alberich-Bayarri
- Quantitative Imaging Biomarkers in Medicine, QUIBIM SL, Edificio Europa, Av. de Aragón, 30, Planta 12, 46021, Valencia, Spain
| | | | - Ignacio Blanquer
- Instituto de Instrumentación para Imagen Molecular (I3M), Universitat Politècnica de València (UPV), c\ Camino de Vera s/n, 46022, Valencia, Spain
| | - J Damian Segrelles
- Instituto de Instrumentación para Imagen Molecular (I3M), Universitat Politècnica de València (UPV), c\ Camino de Vera s/n, 46022, Valencia, Spain
| | - Leonor Cerdá-Alberich
- Biomedical Imaging Research Group (GIBI230), La Fe Health Research Institute, Av. Fernando Abril Martorell 106, Torre E, 46026, Valencia, Spain
| | - Polyxeni Gkontra
- Biomedical Imaging Research Group (GIBI230), La Fe Health Research Institute, Av. Fernando Abril Martorell 106, Torre E, 46026, Valencia, Spain
| | - Barbara Hero
- Department of Pediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - J M García-Aznar
- Multiscale in Mechanical and Biological Engineering, Department of Mechanical Engineering, Universidad de Zaragoza, Zaragoza, Spain.,Aragón Institute of Engineering Research, Zaragoza, Spain
| | - Daniel Keim
- Department of Computer Science, University of Konstanz, Konstanz, Germany
| | - Wolfgang Jentner
- Department of Computer Science, University of Konstanz, Konstanz, Germany
| | | | - Ana Jiménez-Pastor
- Quantitative Imaging Biomarkers in Medicine, QUIBIM SL, Edificio Europa, Av. de Aragón, 30, Planta 12, 46021, Valencia, Spain
| | - Ismael González-Valverde
- Quantitative Imaging Biomarkers in Medicine, QUIBIM SL, Edificio Europa, Av. de Aragón, 30, Planta 12, 46021, Valencia, Spain
| | - Blanca Martínez de Las Heras
- Paediatric Oncology Unit, La Fe University and Polytechnic Hospital, Av. Fernando Abril Martorell 106, Torre G, 2 Floor, 46026, Valencia, Spain
| | - Samira Essiaf
- European Society for Paediatric Oncology, Brussels, Belgium
| | - Dawn Walker
- Department of Computer Science and Insigneo Institute of In Silico Medicine, University of Sheffield, Regent Court, 211 Portobello, Sheffield, UK
| | - Michel Rochette
- Simulation, Modelling and Engineering Software, Ansys Group, Montigny-le-Bretonneux, France
| | - Marian Bubak
- ACC Cyfronet, AGH University of Science and Technology, Sano Centre for Computational Medicine, Nawojki 11, 30-950, Kraków, Poland
| | - Jordi Mestres
- Chemotargets S.L., Carrer de Baldiri Reixac, 4-8 TI05A7 Torre I, planta 5, A7, 08028, Barcelona, Spain
| | - Marco Viceconti
- Department of Industrial Engineering, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Gracia Martí-Besa
- Biomedical Imaging Research Group (GIBI230), La Fe Health Research Institute, Av. Fernando Abril Martorell 106, Torre E, 46026, Valencia, Spain
| | - Adela Cañete
- Paediatric Oncology Unit, La Fe University and Polytechnic Hospital, Av. Fernando Abril Martorell 106, Torre G, 2 Floor, 46026, Valencia, Spain
| | - Paul Richmond
- Department of Computer Science and Insigneo Institute of In Silico Medicine, University of Sheffield, Regent Court, 211 Portobello, Sheffield, UK
| | - Kenneth Y Wertheim
- Department of Computer Science and Insigneo Institute of In Silico Medicine, University of Sheffield, Regent Court, 211 Portobello, Sheffield, UK
| | - Tomasz Gubala
- ACC Cyfronet, AGH University of Science and Technology, Sano Centre for Computational Medicine, Nawojki 11, 30-950, Kraków, Poland
| | - Marek Kasztelnik
- ACC Cyfronet, AGH University of Science and Technology, Sano Centre for Computational Medicine, Nawojki 11, 30-950, Kraków, Poland
| | - Jan Meizner
- ACC Cyfronet, AGH University of Science and Technology, Sano Centre for Computational Medicine, Nawojki 11, 30-950, Kraków, Poland
| | - Piotr Nowakowski
- ACC Cyfronet, AGH University of Science and Technology, Sano Centre for Computational Medicine, Nawojki 11, 30-950, Kraków, Poland
| | | | - Amelia Suárez
- Matical Innovation, Calle de Torija, 5, 28013, Madrid, Spain
| | - Mario Aznar
- Matical Innovation, Calle de Torija, 5, 28013, Madrid, Spain
| | - Giuliana Restante
- Department of Translational Research, University of Pisa, Chair Radiodiagnostica 3, Pisa University Hospital, Via Roma 67, 56126, Pisa, Italy
| | - Emanuele Neri
- Department of Translational Research, University of Pisa, Chair Radiodiagnostica 3, Pisa University Hospital, Via Roma 67, 56126, Pisa, Italy
| |
Collapse
|
166
|
Kommalapati VK, Kumar D, Tangutur AD. Inhibition of JNJ-26481585-mediated autophagy induces apoptosis via ROS activation and mitochondrial membrane potential disruption in neuroblastoma cells. Mol Cell Biochem 2020; 468:21-34. [PMID: 32146648 DOI: 10.1007/s11010-020-03708-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 02/21/2020] [Indexed: 01/14/2023]
Abstract
Neuroblastoma (NB) is the common pediatric tumor of the sympathetic nervous system characterized by poor prognosis. Owing to the challenges such as high tumor heterogeneity, multidrug resistance, minimal residual disease, etc., there is an immediate need for exploring new therapeutic strategies and effective treatments for NB. Herein, in the current study, we explored the unexplored response of NB cells to the second-generation histone deacetylase inhibitor (HDACi) JNJ-26481585(JNJ) and the lysosomotropic agent, Chloroquine (CQ) alone and upon JNJ/CQ treatment as a plausible therapeutic. We identify that while JNJ alone induced autophagy in NB cells, JNJ/CQ treatment decreased the viability and proliferation of NB cells in vitro by switching from autophagy to apoptosis. Further we found that autophagy inhibition by CQ pre-treatment led to the generation of ROS and a decrease in the mitochondrial membrane potential (MMP) that subsequently caused caspase-3-mediated apoptotic cell death in NB cells. Corroborating the above observations, we found that the ROS scavenger N-acetylcysteine (NAC) countered caspase-3 activity and the cells were rescued from apoptosis. Finally, these observations establish that JNJ/CQ treatment resulted in cell death in NB cells by triggering the formation of ROS and disruption of MMP, suggesting that modulation of JNJ-induced autophagy by CQ represents a promising new therapeutic approach in NB.
Collapse
Affiliation(s)
- Vamsi Krishna Kommalapati
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, 500007, India.,Academy of Scientific and Innovative Research, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, Telangana, India
| | - Dinesh Kumar
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, 500007, India.,Academy of Scientific and Innovative Research, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, Telangana, India
| | - Anjana Devi Tangutur
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, 500007, India. .,Academy of Scientific and Innovative Research, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, Telangana, India.
| |
Collapse
|
167
|
Masetti R, Prodi A, Liberatore A, Carfagnini F, Cappelletti E, Leardini D, Pession A, De Carolis E, Cricca M. Occurrence of Albifimbria verrucaria in the Blood of a Female Child With Neuroblastoma. Front Med (Lausanne) 2020; 7:13. [PMID: 32118003 PMCID: PMC7033736 DOI: 10.3389/fmed.2020.00013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 01/13/2020] [Indexed: 11/13/2022] Open
Abstract
We report for the first time the occurrence of a filamentous fungus, Albifimbria verrucaria, in the blood of a pediatric neuroblastoma patient. The Albifimbria genus comprises common soil-inhabiting and saprophytic fungi and has been isolated as a plant pathogen in Northern and Southern Italy. As a human pathogen, A. verrucaria has been implicated in keratitis and can produce trichothecene toxins, which are weakly cytotoxic for mammalian cell lines. A. verrucaria was isolated from blood during the follow-up of a previous coagulase-negative Staphylococcus catheter-related infection. Lung nodules, compatible with fungal infection, had been observed on a CT scan 6 months earlier; they still persist. Possible routes of transmission were considered to be airborne, catheter related, or transfusion dependent, as the patient had undergone platelet and red blood cell transfusions during rescue chemotherapy. No filamentous fungi were isolated from sputum or CVCs. In conclusion, we describe an unprecedented fungemia caused by A. verrucaria and show how an unexpected pathogen may be acquired from the environment by patients at high risk due to immunosuppression. The route of transmission remains unknown.
Collapse
Affiliation(s)
- Riccardo Masetti
- "Lalla Seràgnoli", Hematology-Oncology Unit, Department of Pediatrics, University of Bologna, Bologna, Italy
| | - Antonio Prodi
- Department of Agricultural and Food Sciences, University of Bologna, Bologna, Italy
| | - Andrea Liberatore
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | | | - Eleonora Cappelletti
- Department of Agricultural and Food Sciences, University of Bologna, Bologna, Italy
| | - Davide Leardini
- "Lalla Seràgnoli", Hematology-Oncology Unit, Department of Pediatrics, University of Bologna, Bologna, Italy
| | - Andrea Pession
- "Lalla Seràgnoli", Hematology-Oncology Unit, Department of Pediatrics, University of Bologna, Bologna, Italy
| | - Elena De Carolis
- Institute of Microbiology, IRCCS, Catholic University of the Sacred Heart, Polyclinic University Foundation Agostino Gemelli, Rome, Italy
| | - Monica Cricca
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| |
Collapse
|
168
|
Suh JK, Koh KN, Min SY, Kim YS, Kim H, Im HJ, Namgoong JM, Kim DY, Ahn SD, Lee JJ, Seo JJ. Feasibility and effectiveness of treatment strategy of tandem high-dose chemotherapy and autologous stem cell transplantation in combination with 131 I-MIBG therapy for high-risk neuroblastoma. Pediatr Transplant 2020; 24:e13658. [PMID: 31960542 DOI: 10.1111/petr.13658] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 10/01/2019] [Accepted: 12/18/2019] [Indexed: 12/15/2022]
Abstract
This study was performed to evaluate the safety and effectiveness of tandem HDCT/ASCT combined with targeted radiotherapy using 131 I-MIBG for high-risk neuroblastoma. Patients with high-risk neuroblastoma were treated with 8 to 10 cycles of induction chemotherapy before tandem HDCT/ASCT. Patients received 131 I-MIBG treatment before the second HDCT/ASCT. Local radiotherapy and maintenance therapy were performed after tandem HDCT/ASCT. Between 2012 and 2016, 19 patients were diagnosed with high-risk neuroblastoma in our institution and 18 of them received tandem HDCT/ASCT combined with 131 I-MIBG therapy. For the first HDCT/ASCT regimen, 12 patients received busulfan/melphalan and six patients received melphalan/etoposide/carboplatin. The second HDCT included ThioCy. The median dose of 131 I-MIBG was 17.2 mCi/kg for the first eight patients, while 12 patients in the latter period of the study received reduced dose of 10.7 mCi/kg. The 5-year OS and EFS rates were 79% and 61%, respectively, for all 19 patients with high-risk neuroblastoma, and 83% and 64%, respectively, for 18 patients who completed tandem HDCT/ASCT combined with 131 I-MIBG therapy. Six patients experienced disease relapse and five patients died. Treatment-related mortality was not observed. Among 15 evaluable patients, 11 patients (73%) developed hypothyroidism, six patients (40%) had CKD, and six patients (40%) had growth failure. Hypothyroidism and growth failure were less frequent in patients who received reduced doses of 131 I-MIBG therapy. Tandem HDCT/ASCT combined with HD 131 I-MIBG therapy could be feasible for patients with high-risk neuroblastoma with acceptable toxicity profiles and favorable outcomes.
Collapse
Affiliation(s)
- Jin Kyung Suh
- Divison of Pediatric Hematology/Oncology, Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyung-Nam Koh
- Divison of Pediatric Hematology/Oncology, Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - So Yoon Min
- Divison of Pediatric Hematology/Oncology, Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Young Sun Kim
- Divison of Pediatric Hematology/Oncology, Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyery Kim
- Divison of Pediatric Hematology/Oncology, Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Ho Joon Im
- Department of Pediatric Surgery, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Jung-Man Namgoong
- Department of Pediatric Surgery, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Dae Yeon Kim
- Department of Pediatric Surgery, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Seung Do Ahn
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jong Jin Lee
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jong Jin Seo
- Divison of Pediatric Hematology/Oncology, Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
169
|
Avitabile M, Lasorsa VA, Cantalupo S, Cardinale A, Cimmino F, Montella A, Capasso D, Haupt R, Amoroso L, Garaventa A, Quattrone A, Corrias MV, Iolascon A, Capasso M. Association of PARP1 polymorphisms with response to chemotherapy in patients with high-risk neuroblastoma. J Cell Mol Med 2020; 24:4072-4081. [PMID: 32103589 PMCID: PMC7171401 DOI: 10.1111/jcmm.15058] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/17/2020] [Accepted: 01/27/2020] [Indexed: 12/25/2022] Open
Abstract
The genetic aetiology and the molecular mechanisms that characterize high‐risk neuroblastoma are still little understood. The majority of high‐risk neuroblastoma patients do not take advantage of current induction therapy. So far, one of the main reasons liable for cancer therapeutic failure is the acquisition of resistance to cytotoxic anticancer drugs, because of the DNA repair system of tumour cells. PARP1 is one of the main DNA damage sensors involved in the DNA repair system and genomic stability. We observed that high PARP1 mRNA level is associated with unfavourable prognosis in 3 public gene expression NB patients’ datasets and in 20 neuroblastomas analysed by qRT‐PCR. Among 4983 SNPs in PARP1, we selected two potential functional SNPs. We investigated the association of rs907187, in PARP1 promoter, and rs2048426 in non‐coding region with response chemotherapy in 121 Italian patients with high‐risk NB. Results showed that minor G allele of rs907187 associated with induction response of patients (P = .02) and with decrease PARP1 mRNA levels in NB cell line (P = .003). Furthermore, rs907187 was predicted to alter the binding site of E2F1 transcription factor. Specifically, allele G had low binding affinity with E2F1 whose expression positively correlates with PARP1 expression and associated with poor prognosis of patients with NB. By contrast, we did not find genetic association for the SNP rs2048426. These data reveal rs907187 as a novel potential risk variant associated with the failure of induction therapy for high‐risk NB.
Collapse
Affiliation(s)
- Marianna Avitabile
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples, Italy.,CEINGE Biotecnologie Avanzate, Naples, Italy
| | - Vito Alessandro Lasorsa
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples, Italy.,CEINGE Biotecnologie Avanzate, Naples, Italy
| | | | | | | | | | - Dalila Capasso
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples, Italy.,CEINGE Biotecnologie Avanzate, Naples, Italy
| | - Riccardo Haupt
- UOS Epidemiology, Biostatistics and Committees, Genova, Italy
| | - Loredana Amoroso
- Department of Pediatric Oncology, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Alberto Garaventa
- Department of Pediatric Oncology, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Alessandro Quattrone
- Laboratory of Translational Genomics, Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Maria Valeria Corrias
- Laboratory of Experimental Therapy in Oncology, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Achille Iolascon
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples, Italy.,CEINGE Biotecnologie Avanzate, Naples, Italy
| | - Mario Capasso
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples, Italy.,CEINGE Biotecnologie Avanzate, Naples, Italy.,IRCCS SDN, Naples, Italy
| |
Collapse
|
170
|
Pencil Beam Scanning Proton Therapy for Paediatric Neuroblastoma with Motion Mitigation Strategy for Moving Target Volumes. Clin Oncol (R Coll Radiol) 2020; 32:467-476. [PMID: 32081577 DOI: 10.1016/j.clon.2020.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 11/27/2019] [Accepted: 12/23/2019] [Indexed: 02/04/2023]
Abstract
AIMS More efforts are required to minimise late radiation side-effects for paediatric patients. Pencil beam scanning proton beam therapy (PBS-PT) allows increased sparing of normal tissues while maintaining conformality, but is prone to dose degradation from interplay effects due to respiratory motion. We report our clinical experience of motion mitigation with volumetric rescanning (vRSC) and outcomes of children with neuroblastoma. MATERIALS AND METHODS Nineteen patients with high-risk (n = 16) and intermediate-risk (n = 3) neuroblastoma received PBS-PT. The median age at PBS-PT was 3.5 years (range 1.2-8.6) and the median PBS-PT dose was 21 Gy (relative biological effectiveness). Most children (89%) were treated under general anaesthesia. Seven patients (37%) underwent four-dimensional computed tomography for motion assessment and were treated with vRSC for motion mitigation. RESULTS The mean result of maximum organ motion was 2.7 mm (cranial-caudal), 1.2 mm (left-right), 1.0 mm (anterior-posterior). Four anaesthetised children (21%) showing <5 mm motion had four-dimensional dose calculations (4DDC) to guide the number of vRSC. The mean deterioration or improvement to the planning target volume covered by 95% of the prescribed dose compared with static three-dimensional plans were: 4DDC no vRSC, -0.6%; 2 vRSC, +0.3%; 4 vRSC, +0.3%; and 8 vRSC, +0.1%. With a median follow-up of 14.9 months (range 2.7-49.0) there were no local recurrences. The 2-year overall survival was 94% and distant progression-free survival was 76%. Acute grade 2-4 toxicity was 11%. During the limited follow-up time, no late toxicities were observed. CONCLUSIONS The early outcomes of mainly high-risk patients with neuroblastoma treated with PBS-PT were excellent. With a subset of our cohort undergoing PBS-PT with vRSC we have shown that it is logistically feasible and safe. The clinical relevance of vRSC is debatable in anaesthetised children with small pre-PBS-PT motion of <5 mm.
Collapse
|
171
|
Investigation of the Role of Dinutuximab Beta-Based Immunotherapy in the SIOPEN High-Risk Neuroblastoma 1 Trial (HR-NBL1). Cancers (Basel) 2020; 12:cancers12020309. [PMID: 32013055 PMCID: PMC7072500 DOI: 10.3390/cancers12020309] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/17/2020] [Accepted: 01/25/2020] [Indexed: 11/18/2022] Open
Abstract
To explore the effects of immunotherapy in the International Society of Paediatric Oncology Europe Neuroblastoma Group SIOPEN high-risk neuroblastoma 1 trial (HR-NBL1 trial), two cohorts were studied: one prior to and one after the introduction of dinutuximab beta. All patients received standard induction and high-dose therapy (HDT) with autologous stem cell rescue (ASCR); the local control comprised surgery and radiotherapy to the primary tumour site, followed by isotretinoin. A landmark timepoint of 109 days, resulting from the median time between ASCR and initiation of immunotherapy, was used to define patients’ eligibility in the pre-immunotherapy analysis cohort. Median follow-up was 5.8 years (inter-quartile range (IQR): 4.2–8.2 years) for 844 eligible patients balanced for risk factors, such as age, sex, stage 4, MYCN amplification and response prior to HDT. The five-year event-free and overall survival (95% confidence interval (CI) of 466 patients not receiving immunotherapy was 42% (38–47%) and 50% (46–55%) but was 57% (51–62%) and 64% (59–69%) for 378 patients receiving immunotherapy (p < 0.001). A multivariate analysis identified absence of immunotherapy (p = 0.0002, hazard ratio (HR) 1.573); type of HDT (p = 0.0029, HR 1.431); less than complete response prior to maintenance therapy (p = 0.0043, HR 1.494) and >1 metastatic compartment at diagnosis (p < 0.001, HR 2.665) as risk factors for relapse or progression. Results suggest an important role for dinutuximab beta-based immunotherapy within the treatment concepts applied in HR-NBL1/SIOPEN.
Collapse
|
172
|
Berthold F, Faldum A, Ernst A, Boos J, Dilloo D, Eggert A, Fischer M, Frühwald M, Henze G, Klingebiel T, Kratz C, Kremens B, Krug B, Leuschner I, Schmidt M, Schmidt R, Schumacher-Kuckelkorn R, von Schweinitz D, Schilling FH, Theissen J, Volland R, Hero B, Simon T. Extended induction chemotherapy does not improve the outcome for high-risk neuroblastoma patients: results of the randomized open-label GPOH trial NB2004-HR. Ann Oncol 2020; 31:422-429. [PMID: 32067684 DOI: 10.1016/j.annonc.2019.11.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Long-term survival of high-risk neuroblastoma patients is still below 50% despite intensive multimodal treatment. This trial aimed to address whether the addition of two topotecan-containing chemotherapy courses compared to standard induction therapy improves event-free survival (EFS) of these patients. PATIENTS AND METHODS An open-label, multicenter, prospective randomized controlled trial was carried out at 58 hospitals in Germany and Switzerland. Patients aged 1-21 years with stage 4 neuroblastoma and patients aged 6 months to 21 years with MYCN-amplified tumors were eligible. The primary endpoint was EFS. Patients were randomly assigned to standard induction therapy with six chemotherapy courses or to experimental induction chemotherapy starting with two additional courses of topotecan, cyclophosphamide, and etoposide followed by standard induction chemotherapy (eight courses in total). After induction chemotherapy, all patients received high-dose chemotherapy with autologous hematopoietic stem cell rescue and isotretinoin for consolidation. Radiotherapy was applied to patients with active tumors at the end of induction chemotherapy. RESULTS Of 536 patients enrolled in the trial, 422 were randomly assigned to the control arm (n = 211) and the experimental arm (n = 211); the median follow-up time was 3.32 years (interquartile range 1.65-5.92). At data lock, the 3-year EFS of experimental and control patients was 34% and 32% [95% confidence Interval (CI) 28% to 40% and 26% to 38%; P = 0.258], respectively. Similarly, the 3-year overall survival of the patients did not differ [54% and 48% (95% CI 46% to 62% and 40% to 56%), respectively; P = 0.558]. The response to induction chemotherapy was not different between the arms. The median number of non-fatal toxicities per patient was higher in the experimental group while the median number of toxicities per chemotherapy course was not different. CONCLUSION While the burden for the patients was increased by prolonging the induction chemotherapy and the toxicity, the addition of two topotecan-containing chemotherapy courses did not improve the EFS of high-risk neuroblastoma patients and thus cannot be recommended. CLINICAL TRIALS. GOV NUMBER NCT number 03042429.
Collapse
Affiliation(s)
- F Berthold
- Department of Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany.
| | - A Faldum
- Institute of Medical Statistics and Clinical Research, University of Muenster, Muenster, Germany
| | - A Ernst
- Institute of Medical Statistics and Computational Biology (IMSB), University of Cologne, Cologne, Germany
| | - J Boos
- Department of Pediatric Oncology and Hematology, University of Muenster, Muenster, Germany
| | - D Dilloo
- Department of Pediatric Oncology and Hematology, University of Bonn, Bonn, Germany
| | - A Eggert
- Department of Pediatric Oncology and Hematology, Charité Universitätsmedizin Berlin and Berlin Institute of Health, Berlin, Germany
| | - M Fischer
- Department of Experimental Pediatric Oncology and Center for Molecular Medicine, Medical Faculty, University of Cologne, Cologne, Germany
| | - M Frühwald
- Swabian Children's Cancer Center, Children's Hospital, University Hospital Augsburg, Augsburg, Germany
| | - G Henze
- Department of Pediatric Oncology and Hematology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - T Klingebiel
- Department of Children and Adolescents, University Hospital, Goethe University Frankfurt (Main), Frankfurt am Main, Germany
| | - C Kratz
- Department of Pediatric Oncology and Hematology, Medicinal University, Hannover, Germany
| | - B Kremens
- Department of Pediatric Oncology and Hematology, University of Essen, Essen, Germany
| | - B Krug
- Institute of Diagnostic and Interventional Radiology, University of Cologne, Cologne, Germany
| | - I Leuschner
- Children's Tumor Registry, Institute of Pathology, University of Kiel, Kiel, Germany
| | - M Schmidt
- Department of Nuclear Medicine, University of Cologne, Cologne, Germany
| | - R Schmidt
- Institute of Medical Statistics and Clinical Research, University of Muenster, Muenster, Germany
| | | | - D von Schweinitz
- Department of Pediatric Surgery, University of Munich, Munich, Germany
| | - F H Schilling
- Department of Pediatric Oncology and Hematology, Olgahospital Stuttgart, Stuttgart, Germany
| | - J Theissen
- Department of Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany
| | - R Volland
- Department of Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany
| | - B Hero
- Department of Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany
| | - T Simon
- Department of Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany
| |
Collapse
|
173
|
Plerixafor-aided Mobilization of Peripheral Blood Hematopoietic Stem Cells to Support Subsequent High-dose Chemotherapy After a Prior Autologous Transplant. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2019; 20:e50-e57. [PMID: 31884151 DOI: 10.1016/j.clml.2019.11.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/17/2019] [Accepted: 11/28/2019] [Indexed: 01/22/2023]
Abstract
BACKGROUND An appreciable proportion of patients in need of salvage high-dose chemotherapy (HDC) and autologous peripheral blood stem cell (PBSC) transplantation (PBSCT) fail to mobilize adequate numbers of hematopoietic progenitors, and plerixafor is applied for that purpose. Limited data exist on remobilization of PBSCs in patients who have relapsed after prior HDC + PBSCT. Herein, we report on consecutive patients that had undergone successful prior single or tandem HDC for a variety of malignant neoplasms in our institution, and later required re-mobilization of PBSCs in order to support further HDC cycles. PATIENTS AND METHODS Plerixafor was administered in combination with granulocyte-colony stimulating factor alone, or after mobilizing chemotherapy. Five patients, 2 B-cell non-Hodgkin lymphomas, 1 multiple myeloma, 1 germ-cell tumor, and 1 Ewing sarcoma, having relapsed after prior HDC + PBSCT, were deemed candidates for further cycle(s) of PBSC-supported HDC. Plerixafor was applied in a "just-in-time" strategy after low CD34+ numbers were measured on the first day of anticipated hematopoietic stem cell collection (non-Hodgkin lymphoma, germ-cell tumor, and Ewing sarcoma), or pre-emptively in multiple myeloma. RESULTS Successful collection of adequate PBSCs was achieved in all patients, from 1.8 to 3.8 × 106/kg after a median of 2 (range, 1-3) leukaphereses; 4 of 5 patients underwent subsequent HDC + PBSCT and engrafted after a median of 11 days (range, 9-55 days) and 25 days (range, 17-76 days) for neutrophils and platelets, respectively. CONCLUSION Plerixafor proved effective to mobilize adequate numbers of PBSCs in individual patients with relapsed malignancies after prior single or tandem HDC + PBSCT. These PBSCs could establish sustained multi-lineage hematopoietic engraftment without any sequelae.
Collapse
|
174
|
Merugu S, Chen L, Gavens E, Gabra H, Brougham M, Makin G, Ng A, Murphy D, Gabriel AS, Robinson ML, Wright JH, Burchill SA, Humphreys A, Bown N, Jamieson D, Tweddle DA. Detection of Circulating and Disseminated Neuroblastoma Cells Using the ImageStream Flow Cytometer for Use as Predictive and Pharmacodynamic Biomarkers. Clin Cancer Res 2019; 26:122-134. [DOI: 10.1158/1078-0432.ccr-19-0656] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 08/13/2019] [Accepted: 10/18/2019] [Indexed: 11/16/2022]
|
175
|
Rujkijyanont P, Photia A, Traivaree C, Monsereenusorn C, Anurathapan U, Seksarn P, Sosothikul D, Techavichit P, Sanpakit K, Phuakpet K, Wiangnon S, Chotsampancharoen T, Chainansamit SO, Kanjanapongkul S, Meekaewkunchorn A, Hongeng S. Clinical outcomes and prognostic factors to predict treatment response in high risk neuroblastoma patients receiving topotecan and cyclophosphamide containing induction regimen: a prospective multicenter study. BMC Cancer 2019; 19:961. [PMID: 31619207 PMCID: PMC6796460 DOI: 10.1186/s12885-019-6186-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 09/23/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Neuroblastoma is the most common extra-cranial solid tumor among children. Despite intensive treatment, patients with advanced disease mostly experience dismal outcomes. Here, we proposed the use of topotecan and cyclophosphamide containing induction regimen as an upfront therapy to high risk neuroblastoma patients. METHODS Patients with high risk neuroblastoma undergoing ThaiPOG high risk neuroblastoma protocol from 2016 to 2017 were studied. All patients received 6 cycles of induction regimen consisting of 2 cycles topotecan (1.2 mg/m2/day) and cyclophosphamide (400 mg/m2/day) for 5 days followed by cisplatin (50 mg/m2/day) for 4 days combined with etoposide (200 mg/m2/day) for 3 days on the third and fifth cycles and cyclophosphamide (2100 mg/m2/day) for 2 days combined with doxorubicin (25 mg/m2/day) and vincristine (0.67 mg/m2/day) for 3 days on the fourth and sixth cycles. Treatment response after the 5th cycle before surgery and treatment-related toxicities after each topotecan containing induction cycle were evaluated. Relevant prognostic factors were analyzed to measure the treatment response among those patients. RESULTS In all, 107 high risk neuroblastoma patients were enrolled in the study. After the 5th cycle of induction regimen, the patients achieved complete response (N = 2), very good partial response (N = 40), partial response (N = 46) and mixed response (N = 19). None of the patients experienced stable disease or disease progression. The most significant prognostic factor was type of healthcare system. The most common adverse effect was febrile neutropenia followed by mucositis, diarrhea and elevated renal function. CONCLUSION The topotecan and cyclophosphamide containing induction regimen effectively provides favorable treatment response. The regimen is well tolerated with minimal toxicity among patients with high risk neuroblastoma in Thailand.
Collapse
Affiliation(s)
- Piya Rujkijyanont
- Division of Hematology-Oncology, Department of Pediatrics, Phramongkutklao Hospital and College of Medicine, 315 Ratchawithi Road, Ratchathewi, Bangkok, 10400, Thailand.
| | - Apichat Photia
- Division of Hematology-Oncology, Department of Pediatrics, Phramongkutklao Hospital and College of Medicine, 315 Ratchawithi Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Chanchai Traivaree
- Division of Hematology-Oncology, Department of Pediatrics, Phramongkutklao Hospital and College of Medicine, 315 Ratchawithi Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Chalinee Monsereenusorn
- Division of Hematology-Oncology, Department of Pediatrics, Phramongkutklao Hospital and College of Medicine, 315 Ratchawithi Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Usanarat Anurathapan
- Division of Hematology-Oncology, Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Panya Seksarn
- Division of Hematology-Oncology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Darintr Sosothikul
- Division of Hematology-Oncology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Piti Techavichit
- Division of Hematology-Oncology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Kleebsabai Sanpakit
- Division of Hematology-Oncology, Department of Pediatrics, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kamon Phuakpet
- Division of Hematology-Oncology, Department of Pediatrics, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Surapon Wiangnon
- Faculty of Medicine, Mahasarakham University, Mahasarakham, Thailand
| | - Thirachit Chotsampancharoen
- Division of Hematology-Oncology, Department of Pediatrics, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand
| | | | - Somjai Kanjanapongkul
- Division of Hematology-Oncology, Queen Sirikit National Institute of Child Health, Bangkok, Thailand
| | - Arunotai Meekaewkunchorn
- Division of Hematology-Oncology, Queen Sirikit National Institute of Child Health, Bangkok, Thailand
| | - Suradej Hongeng
- Division of Hematology-Oncology, Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
176
|
Furman WL, Federico SM, McCarville MB, Shulkin BL, Davidoff AM, Krasin MJ, Sahr N, Sykes A, Wu J, Brennan RC, Bishop MW, Helmig S, Stewart E, Navid F, Triplett B, Santana VM, Bahrami A, Anthony G, Yu AL, Hank J, Gillies SD, Sondel PM, Leung WH, Pappo AS. A Phase II Trial of Hu14.18K322A in Combination with Induction Chemotherapy in Children with Newly Diagnosed High-Risk Neuroblastoma. Clin Cancer Res 2019; 25:6320-6328. [PMID: 31601569 DOI: 10.1158/1078-0432.ccr-19-1452] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 07/12/2019] [Accepted: 08/13/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE We sought to evaluate whether combining a humanized antidisialoganglioside mAb (hu14.18K322A) with induction chemotherapy improves early responses and outcomes in children with newly diagnosed high-risk neuroblastoma. PATIENTS AND METHODS We conducted a prospective nonrandomized, single-arm, two-stage, phase II clinical trial. Six courses of induction chemotherapy were coadministered with hu14.18K322A and followed with granulocyte-macrophage colony-stimulating factor (GM-CSF) and low-dose IL2. Consolidation was performed with a busulfan/melphalan preparative regimen. An additional course of hu14.18K322A was administered with parent-derived natural killer cells, when available, during consolidation. Hu14.18K322A, GM-CSF, IL2, and isotretinoin were then administered. Secondary outcomes included reduced tumor volume and semiquantitative 123I-metaiodobenzylguanidine scoring [i.e., Curie scores (CS)] at the end of induction. RESULTS Forty-two patients received hu14.18K322A and induction chemotherapy. This regimen was well tolerated, with continuous-infusion narcotics adjusted to patient tolerance. Partial responses (PR) or better after the first two chemoimmunotherapy courses occurred in 32 patients [76.2%; 95% confidence interval (CI), 60.6-88.0]. This was accompanied by primary tumor volume reductions (median, -76%; range, -100% to 5%). Of 35 patients with stage IV disease who completed induction, 31 had end-of-induction CSs of 2 or less. No patients experienced progression during induction. Two-year event-free survival (EFS) was 85.7% (95% CI, 70.9-93.3). CONCLUSIONS Adding hu14.18K322A to induction chemotherapy produced early PR or better in most patients, reduced tumor volumes, improved CSs at the end of induction, and yielded an encouraging 2-year EFS. These results, if validated in a larger study, may change the standard of care for children with high-risk neuroblastoma.
Collapse
Affiliation(s)
- Wayne L Furman
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee.
| | - Sara M Federico
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | | | - Barry L Shulkin
- Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, Tennessee
| | | | - Matthew J Krasin
- Radiation Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Natasha Sahr
- Biostatistics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - April Sykes
- Biostatistics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jianrong Wu
- Biostatistics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Rachel C Brennan
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | | | - Sara Helmig
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Elizabeth Stewart
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Fariba Navid
- Division of Hematology, Oncology and Bone Marrow Transplant, Children's Hospital of Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Brandon Triplett
- Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Victor M Santana
- Clinical Trials Administration, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Armita Bahrami
- Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Gwendolyn Anthony
- Cancer Center Administration, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Alice L Yu
- University of California, San Diego and Moores Cancer Center and Genomics Research Center, Academia Sinica, Taiwan
| | - Jacquelyn Hank
- Departments of Pediatrics and Human Oncology, University of Wisconsin, Madison, Wisconsin
| | | | - Paul M Sondel
- Departments of Pediatrics and Human Oncology, University of Wisconsin, Madison, Wisconsin
| | - Wing H Leung
- Department of Hematology Oncology, KK Women's and Children's Hospital, Duke-NUS, Singapore
| | - Alberto S Pappo
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| |
Collapse
|
177
|
Tolbert VP, Dvorak CC, Golden C, Vissa M, El-Haj N, Perwad F, Matthay KK, Vo KT. Risk Factors for Transplant-Associated Thrombotic Microangiopathy after Autologous Hematopoietic Cell Transplant in High-Risk Neuroblastoma. Biol Blood Marrow Transplant 2019; 25:2031-2039. [PMID: 31199983 PMCID: PMC9161973 DOI: 10.1016/j.bbmt.2019.06.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/21/2019] [Accepted: 06/05/2019] [Indexed: 12/12/2022]
Abstract
High-risk neuroblastoma has a poor prognosis, and research studies have shown that increasing the intensity of therapy improves outcomes. Autologous hematopoietic cell transplant (aHCT) as consolidation therapy confers a significant survival advantage but is accompanied by significant morbidity. Transplant-associated thrombotic microangiopathy (TA-TMA) is a life-threatening complication caused by endothelial injury that often leads to hemolytic anemia, microthrombotic platelet consumption, and renal injury. Here we investigated the incidence, potential risk factors, and sequelae of TA-TMA in patients with high-risk neuroblastoma. We conducted a retrospective chart review of all patients (n = 141) with neuroblastoma in our institutions who underwent aHCT from 2000 to 2017. Ten patients (7%) developed TA-TMA. The patients in the TA-TMA group were similar to the rest of the subjects in demographics, disease burden, prior therapies, renal function, and timing of transplant. The type of conditioning regimen was the only statistically significant pretransplant variable (P < .001). Six of 15 patients (40%) intended to receive tandem transplants (cyclophosphamide/thiotepa and then carboplatin/etoposide/melphalan (CEM)), 4 of 68 patients (6%) who received conditioning with single CEM, and none of the 56 patients who received busulfan/melphalan were diagnosed with TA-TMA. Patients with TA-TMA were more likely to require intensive care unit transfer, have a longer length of stay in the hospital, and experience a delay or change in their subsequent therapy. In our cohort overall, patients with a delay in therapy after transplant appeared to have a worse overall survival, although the difference was not statistically significant. Because of this high incidence and significant morbidity, we have implemented standardized screening for TA-TMA during and after transplant. We anticipate that screening will lead to earlier intervention and decreased severity of disease.
Collapse
Affiliation(s)
- Vanessa P Tolbert
- Department of Pediatrics, University of California San Francisco School of Medicine and Benioff Children's Hospital, San Francisco, California
| | - Christopher C Dvorak
- Department of Pediatrics, University of California San Francisco School of Medicine and Benioff Children's Hospital, San Francisco, California
| | - Carla Golden
- Division of Hematology/Oncology, University of California San Francisco Benioff Children's Hospital, Oakland, California
| | - Madhav Vissa
- Division of Hematology/Oncology, University of California San Francisco Benioff Children's Hospital, Oakland, California
| | - Nura El-Haj
- Division of Hematology/Oncology, University of California San Francisco Benioff Children's Hospital, Oakland, California
| | - Farzana Perwad
- Department of Pediatrics, University of California San Francisco School of Medicine and Benioff Children's Hospital, San Francisco, California
| | - Katherine K Matthay
- Department of Pediatrics, University of California San Francisco School of Medicine and Benioff Children's Hospital, San Francisco, California
| | - Kieuhoa T Vo
- Department of Pediatrics, University of California San Francisco School of Medicine and Benioff Children's Hospital, San Francisco, California.
| |
Collapse
|
178
|
Hori D, Kobayashi R, Fujita N, Suzumiya J, Suzuki R, Kato K, Kawata T, Fukuda T, Inoue M, Goto H, Hama A, Iwato K, Okumura H, Eto T, Hashii Y, Atsuta Y, Mitsui T. The effectiveness of busulfan-based conditioning regimens for stem cell transplantation against lymphomas in children, adolescents, and young adults in Japan. Pediatr Blood Cancer 2019; 66:e27918. [PMID: 31298481 DOI: 10.1002/pbc.27918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 06/20/2019] [Accepted: 06/21/2019] [Indexed: 11/09/2022]
Abstract
Conditioning regimens for stem cell transplantation (SCT) involving total body irradiation (TBI) are generally preferred over busulfan (BU)-based ones for lymphoid malignancies. However, reports of favorable results using BU against lymphomas have recently emerged. This study sought to compare the effectiveness of BU and TBI regimens for SCT against lymphomas. We retrospectively analyzed 893 lymphoma patients who underwent primary SCT in Japan between 1980 and 2015. The median age of all patients was 18 years (range, 0-30 years) with 589 males, 303 females, and 1 patient whose sex was unknown. Overall survival (OS) was not different between those receiving BU and TBI (P = 0.672). OS in patients receiving autologous SCT was significantly better with BU over TBI regimens (P = 0.038), particularly in children (0-15 years) (P = 0.024). Conversely, OS in adolescents and young adults (AYAs; 16-30 years) receiving allogeneic SCT was significantly worse with BU over TBI regimens (P = 0.035). Overall, BU regiments had comparable effectiveness to TBI conditioning regimens, and, although less effective for AYAs with allogeneic SCT, were particularly more effective than TBI regimens for children who received autologous SCT.
Collapse
Affiliation(s)
- Daiki Hori
- Department of Hematology/Oncology for Children and Adolescents, Sapporo Hokuyu Hospital, Sapporo, Hokkaido, Japan
| | - Ryoji Kobayashi
- Department of Hematology/Oncology for Children and Adolescents, Sapporo Hokuyu Hospital, Sapporo, Hokkaido, Japan
| | - Naoto Fujita
- Department of Pediatrics, Hiroshima Red Cross Hospital and Atomic-bomb Survivors Hospital, Hiroshima, Hiroshima Prefecture, Japan
| | - Junji Suzumiya
- Department of Oncology and Hematology, Shimane University Hospital Innovative Cancer Center, Izumo, Shimane Prefecture, Japan
| | - Ritsuro Suzuki
- Department of Oncology and Hematology, Shimane University Hospital Innovative Cancer Center, Izumo, Shimane Prefecture, Japan
| | - Koji Kato
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Fukuoka Prefecture, Japan
| | - Takahito Kawata
- Department of Hematology, Hyogo Prefectural Amagasaki General Medical Center, Amagasaki, Hyogo Prefecture, Japan
| | - Takahiro Fukuda
- Department of Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| | - Masami Inoue
- Department of Hematology/Oncology, Osaka Women's and Children's Hospital, Izumi, Osaka Prefecture, Japan
| | - Hiroaki Goto
- Division of Hemato-oncology/Regenerative Medicine, Kanagawa Children's Medical Center, Yokohama, Kanagawa Prefecture, Japan
| | - Asahito Hama
- Department of Hematology and Oncology, Children's Medical Center, Japanese Red Cross Nagoya First Hospital, Nagoya, Aichi Prefecture, Japan
| | - Koji Iwato
- Department of Hematology, Hiroshima Red Cross Hospital and Atomic-bomb Survivors Hospital, Hiroshima, Hiroshima Prefecture, Japan
| | - Hirokazu Okumura
- Department of Internal Medicine (Hematology), Toyama Prefectural Central Hospital, Toyama, Toyama Prefecture, Japan
| | - Tetsuya Eto
- Department of Hematology, Hamanomachi Hospital, Fukuoka, Fukuoka Prefecture, Japan
| | - Yoshiko Hashii
- Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka Prefecture, Japan
| | - Yoshiko Atsuta
- Japanese Data Center for Hematopoietic Cell Transplantation, Nagoya, Aichi Prefecture, Japan
| | - Tetsuo Mitsui
- Department of Pediatrics, Yamagata University Hospital, Yamagata, Yamagata Prefecture, Japan
| |
Collapse
|
179
|
Guerreiro F, Zachiu C, Seravalli E, Ribeiro CO, Janssens GO, Ries M, de Senneville BD, Maduro JH, Brouwer CL, Korevaar EW, Knopf AC, Raaymakers BW. Evaluating the benefit of PBS vs. VMAT dose distributions in terms of dosimetric sparing and robustness against inter-fraction anatomical changes for pediatric abdominal tumors. Radiother Oncol 2019; 138:158-165. [DOI: 10.1016/j.radonc.2019.06.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/27/2019] [Accepted: 06/17/2019] [Indexed: 11/16/2022]
|
180
|
Hong CR, Kang HJ, Moon SJ, Oh J, Hong KT, Choi JY, Yu KS, Shin HY. Pharmacokinetics of high-dose carboplatin in children undergoing high-dose chemotherapy and autologous stem cell transplantation with BSA-based dosing. Bone Marrow Transplant 2019; 55:137-146. [PMID: 31462686 DOI: 10.1038/s41409-019-0655-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 05/28/2019] [Accepted: 07/04/2019] [Indexed: 11/09/2022]
Abstract
Body surface area (BSA)-based carboplatin dosing is used in various centers due to practical issues of renal function-based dosing with area under the curve (AUC) measurement. Pharmacokinetic (PK) analysis of high-dose carboplatin was performed in pediatric solid tumor patients undergoing high-dose chemotherapy (HDCT) and autologous stem cell transplantation (ASCT) with BSA-based dosing to calculate the AUCs achieved with this dosing method and to find the correlation between the PK and the renal functions and the adverse events. Carboplatin was administered as once daily intravenous doses at 300, 400, or 500 mg/m2/day over 1 h for 3 or 4 days. On the first and the last day of carboplatin administration, PK samplings were done at 0, 1, 2, and 5 h and only at 0 h on any other days. Mean AUC on the first and the last day were 4.85 ± 0.95 min × mg/mL and 5.27 ± 1.04 min × mg/mL, respectively (n = 23). Overall, negative correlations between the renal functions and the AUCs were mild to moderate, but they were stronger in nephrectomized patients. 51Cr-EDTA clearance decreased with statistical significance with each additional dose of carboplatin (P = 0.020). Optimal high-dose carboplatin dosing method and optimal target AUCs for the different tumors need further analysis.
Collapse
Affiliation(s)
- Che Ry Hong
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea.,Seoul National University Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyoung Jin Kang
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea.,Seoul National University Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seol Ju Moon
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - Jaeseong Oh
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - Kyung Taek Hong
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea.,Seoul National University Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jung Yoon Choi
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea.,Seoul National University Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kyung-Sang Yu
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - Hee Young Shin
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea. .,Seoul National University Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
181
|
Park JR, Kreissman SG, London WB, Naranjo A, Cohn SL, Hogarty MD, Tenney SC, Haas-Kogan D, Shaw PJ, Kraveka JM, Roberts SS, Geiger JD, Doski JJ, Voss SD, Maris JM, Grupp SA, Diller L. Effect of Tandem Autologous Stem Cell Transplant vs Single Transplant on Event-Free Survival in Patients With High-Risk Neuroblastoma: A Randomized Clinical Trial. JAMA 2019; 322:746-755. [PMID: 31454045 PMCID: PMC6714031 DOI: 10.1001/jama.2019.11642] [Citation(s) in RCA: 232] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
IMPORTANCE Induction chemotherapy followed by high-dose therapy with autologous stem cell transplant and subsequent antidisialoganglioside antibody immunotherapy is standard of care for patients with high-risk neuroblastoma, but survival rate among these patients remains low. OBJECTIVE To determine if tandem autologous transplant improves event-free survival (EFS) compared with single transplant. DESIGN, SETTING, AND PARTICIPANTS Patients were enrolled in this randomized clinical trial from November 2007 to February 2012 at 142 Children's Oncology Group centers in the United States, Canada, Switzerland, Australia, and New Zealand. A total of 652 eligible patients aged 30 years or younger with protocol-defined high-risk neuroblastoma were enrolled and 355 were randomized. The final date of follow-up was June 29, 2017, and the data analyses cut-off date was June 30, 2017. INTERVENTIONS Patients were randomized to receive tandem transplant with thiotepa/cyclophosphamide followed by dose-reduced carboplatin/etoposide/melphalan (n = 176) or single transplant with carboplatin/etoposide/melphalan (n = 179). MAIN OUTCOMES AND MEASURES The primary outcome was EFS from randomization to the occurrence of the first event (relapse, progression, secondary malignancy, or death from any cause). The study was designed to test the 1-sided hypothesis of superiority of tandem transplant compared with single transplant. RESULTS Among the 652 eligible patients enrolled, 297 did not undergo randomization because they were nonrandomly assigned (n = 27), ineligible for randomization (n = 62), had no therapy (n = 1), or because of physician/parent preference (n = 207). Among 355 patients randomized (median diagnosis age, 36.1 months; 152 [42.8%] female), 297 patients (83.7%) completed the study and 21 (5.9%) were lost to follow-up after completing protocol therapy. Three-year EFS from the time of randomization was 61.6% (95% CI, 54.3%-68.9%) in the tandem transplant group and 48.4% (95% CI, 41.0%-55.7%) in the single transplant group (1-sided log-rank P=.006). The median (range) duration of follow-up after randomization for 181 patients without an event was 5.6 (0.6-8.9) years. The most common significant toxicities following tandem vs single transplant were mucosal (11.7% vs 15.4%) and infectious (17.9% vs 18.3%). CONCLUSIONS AND RELEVANCE Among patients aged 30 years or younger with high-risk neuroblastoma, tandem transplant resulted in a significantly better EFS than single transplant. However, because of the low randomization rate, the findings may not be representative of all patients with high-risk neuroblastoma. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT00567567.
Collapse
Affiliation(s)
- Julie R. Park
- Department of Pediatrics, Seattle Children’s Hospital, Seattle, Washington
- University of Washington, Seattle
| | - Susan G. Kreissman
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina
| | - Wendy B. London
- Department of Pediatrics, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts
| | - Arlene Naranjo
- Department of Biostatistics, University of Florida, Children’s Oncology Group Statistics and Data Center, Gainesville
| | | | - Michael D. Hogarty
- Department of Pediatrics Children’s Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Sheena C. Tenney
- Department of Biostatistics, University of Florida, Children’s Oncology Group Statistics and Data Center, Gainesville
| | - Daphne Haas-Kogan
- Department of Radiation Oncology, Dana Farber/Brigham and Women’s Cancer Center and Harvard Medical School, Boston, Massachusetts
| | - Peter John Shaw
- Bone Marrow Transplant, Children's Hospital at Westmead, Sydney, Australia
| | | | - Stephen S. Roberts
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, Manhattan, New York
| | - James Duncan Geiger
- Section of Pediatric Surgery, CS Mott Children’s Hospital, Michigan Medicine, Ann Arbor
| | - John J. Doski
- Departments of Surgery and Pediatrics, UT Health San Antonio, San Antonio, Texas
| | - Stephan D. Voss
- Department of Radiology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - John M. Maris
- Department of Pediatrics Children’s Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Stephan A. Grupp
- Department of Pediatrics Children’s Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Lisa Diller
- Department of Pediatrics, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
182
|
Bagatell R, Irwin MS. Tandem Transplant for High-Risk Neuroblastoma: Next Steps in the Era of Precision Medicine. JAMA 2019; 322:729-731. [PMID: 31454025 DOI: 10.1001/jama.2019.11641] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Rochelle Bagatell
- Perelman School of Medicine, Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia
| | - Meredith S Irwin
- Division of Hematology/Oncology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
183
|
Linke C, Tragiannidis A, Ahlmann M, Fröhlich B, Wältermann M, Burkhardt B, Rossig C, Groll AH. Epidemiology and management burden of invasive fungal infections after autologous hematopoietic stem cell transplantation: 10-year experience at a European Pediatric Cancer Center. Mycoses 2019; 62:954-960. [PMID: 31332851 DOI: 10.1111/myc.12968] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 06/24/2019] [Accepted: 07/17/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Autologous hematopoietic stem cell transplantation (HSCT) carries risks of infectious morbidity. We analysed epidemiology and management burden associated with invasive fungal diseases (IFDs) in children and adolescents undergoing autologous HSCT. METHODS In a retrospective, single-centre observational study, epidemiology and management burden associated with IFDs were analysed in all paediatric cancer patients who underwent autologous HSCT between 2005 and 2014. Clinical, radiographic and microbiological data were assessed up to 100 days post-transplant. The primary endpoint was the incidence of proven, probable and possible IFDs. Further endpoints included the use of systemic antifungal agents for prevention and management of IFDs; infectious and non-infectious comorbidities; and survival until day + 100. RESULTS Of 95 patients (median age: 8 years; r, 0.75-20) underwent 103 HSCT procedures for solid tumours (92) or lymphoma (11). Primary antifungal prophylaxis was administered in 49 procedures (47.5%). No single case of proven/probable IFD was diagnosed. Nine cases (8.7%) fulfilled criteria of possible pulmonary mould infection and received treatment for a median of 14 days (r, 7-35). In an additional 12 procedures, empiric antifungal therapy with mould active agents was given for a median of 8 days (r, 3-105). Microbiologically documented non-fungal infections were observed in 17 procedures, and five patients were transferred to the ICU. There was one death from biopsy documented toxic endothelial damage at day 83 post-transplant. CONCLUSIONS Autologous HSCT for solid tumours or lymphoma was associated with low morbidity from IFDs. However, utilisation of systemic antifungal agents for prevention and management of suspected IFDs was considerable.
Collapse
Affiliation(s)
- Christina Linke
- Infectious Disease Research Program, University Children's Hospital Münster, Münster, Germany.,Center for Bone Marrow Transplantation, University Children's Hospital Münster, Münster, Germany.,Hematology Oncology Unit, 2nd Department of Pediatrics, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Athanasios Tragiannidis
- Infectious Disease Research Program, University Children's Hospital Münster, Münster, Germany.,Center for Bone Marrow Transplantation, University Children's Hospital Münster, Münster, Germany.,Hematology Oncology Unit, 2nd Department of Pediatrics, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Martina Ahlmann
- Center for Bone Marrow Transplantation, University Children's Hospital Münster, Münster, Germany.,Hematology Oncology Unit, 2nd Department of Pediatrics, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Birgit Fröhlich
- Center for Bone Marrow Transplantation, University Children's Hospital Münster, Münster, Germany.,Hematology Oncology Unit, 2nd Department of Pediatrics, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Maria Wältermann
- Center for Bone Marrow Transplantation, University Children's Hospital Münster, Münster, Germany.,Hematology Oncology Unit, 2nd Department of Pediatrics, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Birgit Burkhardt
- Center for Bone Marrow Transplantation, University Children's Hospital Münster, Münster, Germany.,Hematology Oncology Unit, 2nd Department of Pediatrics, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Claudia Rossig
- Center for Bone Marrow Transplantation, University Children's Hospital Münster, Münster, Germany.,Hematology Oncology Unit, 2nd Department of Pediatrics, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Andreas H Groll
- Infectious Disease Research Program, University Children's Hospital Münster, Münster, Germany.,Center for Bone Marrow Transplantation, University Children's Hospital Münster, Münster, Germany.,Hematology Oncology Unit, 2nd Department of Pediatrics, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
184
|
Ollé Hurtado M, Wolbert J, Fisher J, Flutter B, Stafford S, Barton J, Jain N, Barone G, Majani Y, Anderson J. Tumor infiltrating lymphocytes expanded from pediatric neuroblastoma display heterogeneity of phenotype and function. PLoS One 2019; 14:e0216373. [PMID: 31398192 PMCID: PMC6688820 DOI: 10.1371/journal.pone.0216373] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 04/18/2019] [Indexed: 01/06/2023] Open
Abstract
Adoptive transfer of ex vivo expanded tumor infiltrating lymphocytes (TILs) has led to clinical benefit in some patients with melanoma but has not demonstrated convincing efficacy in other solid cancers. Whilst the presence of TILs in many types of cancer is often associated with better clinical prognosis, their function has not been systematically evaluated across cancer types. Responses to immunological checkpoint inhibitors in a wide range of cancers, including those for which adoptive transfer of expanded TILs has not shown clinical benefit, has clearly delineated a number of tumor type associated with tumor-reactive lymphocytes capable of effecting tumor remissions. Neuroblastoma is an aggressive childhood solid cancer in which immunotherapy with GD2-directed antibodies confers a proven survival advantage through incompletely understood mechanisms. We therefore evaluated the feasibility of ex vivo expansion of TILs from freshly resected neuroblastoma tumors and the potential therapeutic utility of TIL expansions. TILs were successfully expanded from both tumor biopsies or resections. Significant numbers of NKT and γδT cells were identified alongside the mixed population of cytotoxic (CD8+) and helper (CD4+) T cells of both effector and central memory phenotypes. Isolated TILs were broadly non-reactive against autologous tumor and neuroblastoma cell lines, so enhancement of neuroblastoma killing was attained by transducing TILs with a second-generation chimeric antigen receptor (CAR) targeting GD2. CAR-TILs demonstrated antigen-specific cytotoxicity against tumor cell lines. This study is the first to show reproducible expansion of TILs from pediatric neuroblastoma, the high proportion of innate-like lymphocytes, and the feasibility to use CAR-TILs therapeutically.
Collapse
Affiliation(s)
- Marina Ollé Hurtado
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, England, United Kingdom
| | - Jolien Wolbert
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, England, United Kingdom
| | - Jonathan Fisher
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, England, United Kingdom
| | - Barry Flutter
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, England, United Kingdom
| | - Sian Stafford
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, England, United Kingdom
| | - Jack Barton
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, England, United Kingdom
| | - Neha Jain
- Department of Oncology, Great Ormond Street Hospital, London, England, United Kingdom
| | - Giuseppe Barone
- Department of Oncology, Great Ormond Street Hospital, London, England, United Kingdom
| | - Yvonne Majani
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, England, United Kingdom
| | - John Anderson
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, England, United Kingdom
- Department of Oncology, Great Ormond Street Hospital, London, England, United Kingdom
- * E-mail:
| |
Collapse
|
185
|
Twist CJ, Schmidt ML, Naranjo A, London WB, Tenney SC, Marachelian A, Shimada H, Collins MH, Esiashvili N, Adkins ES, Mattei P, Handler M, Katzenstein H, Attiyeh E, Hogarty MD, Gastier-Foster J, Wagner E, Matthay KK, Park JR, Maris JM, Cohn SL. Maintaining Outstanding Outcomes Using Response- and Biology-Based Therapy for Intermediate-Risk Neuroblastoma: A Report From the Children's Oncology Group Study ANBL0531. J Clin Oncol 2019; 37:3243-3255. [PMID: 31386611 DOI: 10.1200/jco.19.00919] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
PURPOSE The primary objective of the Children's Oncology Group study ANBL0531 (ClinicalTrials.gov identifier: NCT00499616) was to reduce therapy for subsets of patients with intermediate-risk neuroblastoma using a biology- and response-based algorithm to assign treatment duration while maintaining a 3-year overall survival (OS) of 95% or more for the entire cohort. PATIENTS AND METHODS Children younger than age 12 years with intermediate-risk stage 2A/2B or stage 3 tumors with favorable histology; infants younger than age 365 days with stage 3, 4 or 4S disease; and toddlers from 365 to younger than 547 days with favorable histology, hyperdiploid stage 4, or unfavorable histology stage 3 tumors were eligible. Patients with MYCN-amplified tumors were excluded. Patients were assigned to initially receive two (group 2), four (group 3), or eight (group 4) cycles of chemotherapy with or without surgery on the basis of prognostic markers, including allelic status of chromosomes 1p and 11q; ultimate duration of therapy was determined by overall response. RESULTS Between 2007 and 2011, 404 evaluable patients were enrolled. Compared with legacy Children's Oncology Group studies, subsets of patients had a reduction in treatment. The 3-year event-free survival and OS rates were 83.2% (95% CI, 79.4% to 87.0%) and 94.9% (95% CI, 92.7% to 97.2%), respectively. Infants with stage 4 tumors with favorable biology (n = 61) had superior 3-year event-free survival compared with patients with one or more unfavorable biologic features (n = 47; 86.9% [95% CI, 78.3% to 95.4%] v 66.8% [95% CI, 53.1% to 80.6%]; P = .02), with a trend toward OS advantage (95.0% [95% CI, 89.5% to 100%] v 86.7% [95% CI, 76.6% to 96.7%], respectively; P = .08). OS for patients with localized disease was 100%. CONCLUSION Excellent survival was achieved with this treatment algorithm, with reduction of therapy for subsets of patients. More-effective treatment strategies still are needed for infants with unfavorable biology stage 4 disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Peter Mattei
- Children's Hospital of Philadelphia, Philadelphia, PA
| | | | | | - Edward Attiyeh
- Children's Hospital of Philadelphia, Philadelphia, PA.,Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Michael D Hogarty
- Children's Hospital of Philadelphia, Philadelphia, PA.,Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Julie Gastier-Foster
- Nationwide Children's Hospital, Columbus, OH.,The Ohio State University College of Medicine, Columbus, OH
| | | | - Katherine K Matthay
- University of California, San Francisco, School of Medicine, and UCSF Benioff Children's Hospital, San Francisco, CA
| | | | - John M Maris
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | | |
Collapse
|
186
|
Campbell K, Shyr D, Bagatell R, Fischer M, Nakagawara A, Nieto AC, Brodeur GM, Matthay KK, London WB, DuBois SG. Comprehensive evaluation of context dependence of the prognostic impact of MYCN amplification in neuroblastoma: A report from the International Neuroblastoma Risk Group (INRG) project. Pediatr Blood Cancer 2019; 66:e27819. [PMID: 31115156 PMCID: PMC7323455 DOI: 10.1002/pbc.27819] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/27/2019] [Accepted: 04/29/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND MYCN amplification (MYCN-A) is an established adverse prognostic factor in neuroblastoma. The extent to which the prognostic impact of MYCN-A depends on other factors has not been fully characterized. PATIENTS AND METHODS Using the International Neuroblastoma Risk Group database, we constructed Cox models of overall survival (OS) to obtain hazard ratios of the effect of MYCN-A within subgroups defined by other prognostic factors. Cox models assessed the degree to which the prognostic impact of MYCN-A was modulated by each other covariate. We used absolute hazard ratio (HR) differences to construct classification trees to identify subgroups with greatest differential prognostic effect of MYCN-A. RESULTS In a cohort of 6223 patients with known MYCN status, the OS hazard ratio associated with MYCN-A was 6.3 (95% confidence interval 5.7-7.0, P < .001). Age at diagnosis conferred the largest HR absolute difference for MYCN-A between subgroups (HR absolute difference 16.6; HRs for MYCN-A of 19.6 for <18 months, 3.0 for ≥18 months). MYCN-A remained significantly prognostic of OS after controlling for other factors, abrogating their prognostic strength. Patients whose outcome was most impacted by MYCN status were those who were <18 months, had high mitosis karrhyohexis index (MKI) and low ferritin. CONCLUSION The prognostic strength of MYCN-A varies depending on which patient subgroup defined by other neuroblastoma risk factors is examined, with greatest strength in patients with otherwise favorable features. MYCN-A has little effect within some subgroups, aiding clinical decision-making if MYCN status cannot be assessed. Subgroups where MYCN-A has large effect may be prioritized for agents targeting Myc family proteins.
Collapse
Affiliation(s)
- Kevin Campbell
- Department of Pediatric Oncology, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center and Harvard Medical School, Boston, Massachusetts
| | - Derek Shyr
- Department of Pediatric Oncology, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center and Harvard Medical School, Boston, Massachusetts
| | - Rochelle Bagatell
- Department of Pediatrics, Children’s Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Matthias Fischer
- Department of Pediatric Oncology, Children’s Hospital and Center for Molecular Medicine Cologne, University of Cologne, Köln, Germany
| | - Akira Nakagawara
- SAGA Heavy Ion Medical Accelerator in Tosu (HIMAT) Foundation, Tosu, Japan
| | | | - Garrett M. Brodeur
- Department of Pediatrics, Children’s Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Katherine K. Matthay
- UCSF School of Medicine and UCSF Benioff Children’s Hospital, San Francisco, California
| | - Wendy B. London
- Department of Pediatric Oncology, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center and Harvard Medical School, Boston, Massachusetts
| | - Steven G. DuBois
- Department of Pediatric Oncology, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
187
|
Braunstein SE, London WB, Kreissman SG, Villablanca JG, Davidoff AM, DeSantes K, Castleberry RP, Murray K, Diller L, Matthay K, Cohn SL, Shulkin B, von Allmen D, Parisi MT, Van Ryn CC, Park JR, Quaglia MPL, Haas-Kogan DA. Role of the extent of prophylactic regional lymph node radiotherapy on survival in high-risk neuroblastoma: A report from the COG A3973 study. Pediatr Blood Cancer 2019; 66:e27736. [PMID: 30968542 PMCID: PMC7281832 DOI: 10.1002/pbc.27736] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 02/18/2019] [Accepted: 03/09/2019] [Indexed: 12/13/2022]
Abstract
PURPOSE Neuroblastoma is the most common extracranial solid pediatric malignancy, with poor outcomes in high-risk disease. Standard treatment approaches employ an increasing array of aggressive multimodal therapies, of which local control with surgery and radiotherapy remains a backbone; however, the benefit of broad regional nodal irradiation remains controversial. We analyzed centrally reviewed radiation therapy data from patients enrolled on COG A3973 to evaluate the impact of primary site irradiation and the extent of regional nodal coverage stratified by extent of surgical resection. METHODS Three hundred thirty high-risk neuroblastoma patients with centrally reviewed radiotherapy plans were analyzed. Outcome was evaluated by the extent of nodal irradiation. For the 171 patients who also underwent surgery (centrally reviewed), outcome was likewise analyzed according to the extent of resection. Overall survival (OS), event-free survival (EFS), and cumulative incidence of local progression (CILP) were examined by Kaplan-Meier, log-rank test (EFS, OS), and Grey test (CILP). RESULTS The five-year CILP, EFS, and OS for all 330 patients receiving radiotherapy on A3973 were 8.5% ± 1.5%, 47.2% ± 3.0%, and 59.7% ± 3.0%, respectively. There were no significant differences in outcomes based on the extent of lymph node irradiation regardless of the degree of surgical resection (< 90% or ≥90%). CONCLUSION Although local control remains a significant component of treatment of high-risk neuroblastoma, our results suggest there is no benefit of extensive lymph node irradiation, irrespective of the extent of surgical resection preceding stem cell transplant.
Collapse
Affiliation(s)
| | - Wendy B. London
- Department of Pediatric Oncology/Hematology, Biostatistics Division, Dana Farber/Children’s Hospital Cancer Center
| | | | - Judith G. Villablanca
- Department of Pediatrics, Keck School of Medicine, University of Southern California
| | - Andrew M. Davidoff
- Department of Surgery, Pediatrics Division, St. Jude’s Children’s Research Hospital
| | | | | | - Kevin Murray
- Department of Pediatrics, University of Louisville
| | - Lisa Diller
- Department of Pediatric Oncology/Hematology, Dana Farber/Children’s Hospital Cancer Center
| | - Katherine Matthay
- Department of Pediatric Hematology-Oncology, University of California, San Francisco
| | - Susan L. Cohn
- Department of Pediatrics, Section of Hematology/Oncology, University of Chicago
| | - Barry Shulkin
- Department of Diagnostic Imaging, Pediatrics Division, St. Jude’s Children’s Research Hospital
| | | | | | - C. Collin Van Ryn
- Department of Biostatistics, University of Florida, College of Public Health
| | | | | | | |
Collapse
|
188
|
Chui CH, Lee A. Management of thoracoabdominal neuroblastoma: a 13-year experience. WORLD JOURNAL OF PEDIATRIC SURGERY 2019; 2:e000055. [DOI: 10.1136/wjps-2019-000055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
BackgroundThoracoabdominal neuroblastoma is a unique surgical challenge. We reviewed our experience focusing on disease patterns and corresponding surgical approaches.MethodsAmong 310 patients in our neuroblastoma database, 30 (9.7%) had thoracoabdominal neuroblastoma. Patients’ clinical charts were reviewed and analyzed. Two disease patterns were identified: solitary thoracoabdominal tumor (group A, n=15) and multifocal tumors in thorax and abdomen (group B, n=15). Operative approaches were categorized based on routes of surgical access.ResultsThirty patients with average age of 4.1 (range 0.8–12.8) years were studied. All received preoperative chemotherapy. Among 15 group A patients, four were stage 3 intermediate risk (IR) and 11 were stage 4 high risk (HR). Surgical approaches included single-incision thoracoabdominal approach (n=10), laparotomy-cum-transdiaphragmatic approach (n=3), and laparotomy-cum-thoracotomy approach (n=2). One patient had 10% residual disease and the rest achieved gross total resection. Postoperative complications included chylous ascites (n=3), intestinal obstruction (n=3), pneumonia (n=1), spinal cord infarction (n=1) scoliosis (n=2) and thoracoabdominal nerves palsy (n=3). Among 15 group B patients, all were stage 4 with five IR and 10 HR. Thoracic components were found in the posterior mediastinum (n=7), superior mediastinum (n=4), middle mediastinum (n=1), parietal pleura (n=3) and lungs (n=3). Surgical approaches included multi-incision laparotomy-cum-thoracotomy (n=14) and laparotomy-cum-transdiaphragmatic approach (n=1). Gross total resection was achieved in all surgeries. Postoperative complications included chylous ascites (n=3). Overall, all nine IR patients survived without evidence of disease and 9 (42.8%) HR patients died of disease. There was no perioperative mortality.ConclusionSurgical resection of thoracoabdominal neuroblastoma is feasible and safe. Despite its complexity, thoracoabdominal neuroblastoma has comparable treatment outcomes when compared with single-compartmental disease.
Collapse
|
189
|
Adamson PC, Veal GJ, Womer RB, Meany HJ, Bernhardt MB, Frazier AL, Balis FM. Fundamental problems with pediatric adaptive dosing of carboplatin using nuclear-medicine-based estimates of renal function. Pediatr Blood Cancer 2019; 66:e27672. [PMID: 30767382 DOI: 10.1002/pbc.27672] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 12/17/2018] [Accepted: 02/04/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Carboplatin is often adaptively dosed based on glomerular filtration rate (GFR), usually estimated by nuclear medicine tests. At least five pediatric adaptive dosing formulas have been developed. In an effort to standardize dosing in Children's Oncology Group protocols, we explored methodologic variation in GFR estimation and adaptive-dosing formula performance. PROCEDURE Nuclear medicine GFR data from published series of ≥100 children with cancer were compared. Data from patients for whom body surface area, weight, GFR, and tracer half-life were available were used to compare formulas. RESULTS Differences in methods used to estimate GFR in children with cancer resulted in highly variable population results, with median GFRs ranging from 96 to 150 mL/min/1.73m2 . The choice of adaptive formula had a major impact on calculated dose. When targeting an area under the curve of 7.9 mg/mL • min, the median difference between the formula yielding the lowest and highest carboplatin dose for individual subjects was 289 (range 96-1 737) mg/m2 . CONCLUSIONS Wide variation in GFR obtained with nuclear-medicine-based tests in children with cancer primarily results from systematic methodologic errors. Formulas for calculating carboplatin dose produce additional and substantial variation that may place children with cancer at unnecessary risk for excessive toxicity or underdosing. These findings indicate a need for the development of a uniform, validated method for GFR determination in children that should be utilized in all centers. Currently, adaptive dosing of carboplatin based on GFR has serious limitations and in most clinical settings should arguably not be used in place of body-surface-area-based dosing.
Collapse
Affiliation(s)
- Peter C Adamson
- Division of Oncology, The Children's Hospital of Philadelphia & Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Gareth J Veal
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - Richard B Womer
- Division of Oncology, The Children's Hospital of Philadelphia & Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Holly J Meany
- Department of Hematology/Oncology, Children's National Medical Center, Washington, District of Columbia
| | | | - A Lindsay Frazier
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts
| | - Frank M Balis
- Division of Oncology, The Children's Hospital of Philadelphia & Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
190
|
Wawrzyniak-Dzierżek E, Gajek K, Rybka B, Ryczan-Krawczyk R, Węcławek-Tompol J, Raciborska A, Mielcarek-Siedziuk M, Frączkiewicz J, Salamonowicz M, Kałwak K, Rosa M, Ślęzak A, Ussowicz M. Feasibility and Safety of Treosulfan, Melphalan, and Thiotepa-Based Megachemotherapy with Autologous or Allogeneic Stem Cell Transplantation in Heavily Pretreated Children with Relapsed or Refractory Neuroblastoma. Biol Blood Marrow Transplant 2019; 25:1792-1797. [PMID: 31085306 DOI: 10.1016/j.bbmt.2019.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 05/04/2019] [Accepted: 05/06/2019] [Indexed: 12/22/2022]
Abstract
The prognosis of resistant or relapsing children with neuroblastoma remains very poor, and the search for new therapies is ongoing. In this analysis, we assessed the toxicity of a treosulfan, melphalan, and thiotepa (TMT) regimen in 17 children with recurrent or refractory neuroblastoma who underwent stem cell transplantation (SCT). For allogeneic SCT, fludarabine and antithymocyte globulin were added. The stem cell source was autologous in 8 patients, haploidentical in 8 patients, and a matched unrelated donor in 1 patient. The reported nonhematologic toxicities included grade 3 mucositis, grade 1 to 3 hypertransaminasemia, and in 3 patients, veno-occlusive disease. No neurologic, cardiac, or dermatologic toxicities were observed. The probability of overall survival (OS) in patients with primary resistance was superior to that in patients with relapsed disease (100% versus 22.6%; P = .046). Post-transplantation dinutuximab beta immunotherapy was associated with superior 5-year OS (66.7% versus 11.4%; P = .0007). The use of an allogeneic donor, previous autologous SCT with busulfan and melphalan, and pretreatment with high-dose metaiodobenzylguanidine therapy demonstrated no effect on outcomes. In 4 patients, TMT megatherapy alone was enough to achieve complete remission. The TMT conditioning regimen was well tolerated in heavily pretreated patients with neuroblastoma. The manageable toxicity and addition of new anticancer drugs with optional post-SCT immunotherapy or chemotherapy support further trials with the TMT regimen in patients with neuroblastoma.
Collapse
Affiliation(s)
- Elżbieta Wawrzyniak-Dzierżek
- Department of Pediatric Bone Marrow Transplantation, Oncology, and Hematology, Wroclaw Medical University, Wroclaw, Poland
| | - Kornelia Gajek
- Department of Pediatric Bone Marrow Transplantation, Oncology, and Hematology, Wroclaw Medical University, Wroclaw, Poland
| | - Blanka Rybka
- Department of Pediatric Bone Marrow Transplantation, Oncology, and Hematology, Wroclaw Medical University, Wroclaw, Poland
| | - Renata Ryczan-Krawczyk
- Department of Pediatric Bone Marrow Transplantation, Oncology, and Hematology, Wroclaw Medical University, Wroclaw, Poland
| | - Jadwiga Węcławek-Tompol
- Department of Pediatric Bone Marrow Transplantation, Oncology, and Hematology, Wroclaw Medical University, Wroclaw, Poland
| | - Anna Raciborska
- Department of Oncology and Surgical Oncology for Children and Youth, Institute of Mother and Child, Warsaw, Poland
| | - Monika Mielcarek-Siedziuk
- Department of Pediatric Bone Marrow Transplantation, Oncology, and Hematology, Wroclaw Medical University, Wroclaw, Poland
| | - Jowita Frączkiewicz
- Department of Pediatric Bone Marrow Transplantation, Oncology, and Hematology, Wroclaw Medical University, Wroclaw, Poland
| | - Małgorzata Salamonowicz
- Department of Pediatric Bone Marrow Transplantation, Oncology, and Hematology, Wroclaw Medical University, Wroclaw, Poland
| | - Krzysztof Kałwak
- Department of Pediatric Bone Marrow Transplantation, Oncology, and Hematology, Wroclaw Medical University, Wroclaw, Poland
| | - Monika Rosa
- Department of Pediatric Bone Marrow Transplantation, Oncology, and Hematology, Wroclaw Medical University, Wroclaw, Poland
| | - Aleksandra Ślęzak
- Department of Pediatric Bone Marrow Transplantation, Oncology, and Hematology, Wroclaw Medical University, Wroclaw, Poland
| | - Marek Ussowicz
- Department of Pediatric Bone Marrow Transplantation, Oncology, and Hematology, Wroclaw Medical University, Wroclaw, Poland.
| |
Collapse
|
191
|
Morgenstern DA, Bagatell R, Cohn SL, Hogarty MD, Maris JM, Moreno L, Park JR, Pearson AD, Schleiermacher G, Valteau-Couanet D, London WB, Irwin MS. The challenge of defining "ultra-high-risk" neuroblastoma. Pediatr Blood Cancer 2019; 66:e27556. [PMID: 30479064 DOI: 10.1002/pbc.27556] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/08/2018] [Accepted: 10/27/2018] [Indexed: 12/17/2022]
Abstract
Given the biological and clinical heterogeneity of neuroblastoma, risk stratification is vital to determining appropriate treatment. Historically, most patients with high-risk neuroblastoma (HR-NBL) have been treated uniformly without further stratification. Attempts have been made to identify factors that can be used to risk stratify these patients and to characterize an "ultra-high-risk" (UHR) subpopulation with particularly poor outcome. However, among published data, there is a lack of consensus in the definition of the UHR population and heterogeneity in the endpoints and statistical methods used. This review summarizes our current understanding of stratification of HR-NBL and discusses the complex issues in defining UHR neuroblastoma.
Collapse
Affiliation(s)
| | - Rochelle Bagatell
- Children's Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | | | - Michael D Hogarty
- Children's Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - John M Maris
- Children's Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Lucas Moreno
- Hospital Universitario Niño Jesus, Madrid, Spain
| | - Julie R Park
- Seattle Children's Hospital and University of Washington School of Medicine, Seattle, Washington
| | - Andrew D Pearson
- Institute of Cancer Research and Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, Surrey, UK
| | | | | | - Wendy B London
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts
| | - Meredith S Irwin
- Hospital for Sick Children and University of Toronto, Toronto, Canada
| |
Collapse
|
192
|
Carfilzomib enhances cisplatin-induced apoptosis in SK-N-BE(2)-M17 human neuroblastoma cells. Sci Rep 2019; 9:5039. [PMID: 30911132 PMCID: PMC6434076 DOI: 10.1038/s41598-019-41527-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 03/06/2019] [Indexed: 12/14/2022] Open
Abstract
Neuroblastoma is a solid malignant tumor of the sympathetic nervous system, which accounts for 8–10% of childhood cancers. Considering the overall high risk and poor prognosis associated with neuroblastoma, effective therapeutics should be developed to improve patient survival and quality of life. A recent study showed that a proteasome inhibitor, carfilzomib (CFZ), reduced cell viability of SK-N-BE(2)-M17 neuroblastoma cells. Therefore, we investigated the molecular mechanisms by which CFZ lower the cell viability of neuroblastoma cells. CFZ reduced cell viability via cell cycle arrest at G2/M and apoptosis, which involved caspase activation (caspases-8, 9, 4, and 3), endoplasmic reticulum stress, reactive oxygen species production, mitochondrial membrane potential loss, and autophagy in a dose- and time-dependent manner. The effect of CFZ was additive to that of cisplatin (Cis), a well-known chemotherapeutic drug, in terms of cell viability reduction, cell cycle arrest, and apoptosis. Importantly, the additive effect of CFZ was maintained in Cis-resistant neuroblastoma cells. These results suggest that CFZ can be used in combination therapy for patients with neuroblastoma to overcome the resistance and adverse side effects of Cis.
Collapse
|
193
|
Guerreiro F, Koivula L, Seravalli E, Janssens GO, Maduro JH, Brouwer CL, Korevaar EW, Knopf AC, Korhonen J, Raaymakers BW. Feasibility of MRI-only photon and proton dose calculations for pediatric patients with abdominal tumors. Phys Med Biol 2019; 64:055010. [PMID: 30669135 DOI: 10.1088/1361-6560/ab0095] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The purpose of this study was to develop a method enabling synthetic computed tomography (sCT) generation of the whole abdomen using magnetic resonance imaging (MRI) scans of pediatric patients with abdominal tumors. The proposed method relies on an automatic atlas-based segmentation of bone and lungs followed by an MRI intensity to synthetic Hounsfield unit conversion. Separate conversion algorithms were used for bone, lungs and soft-tissue. Rigidly registered CT and T2-weighted MR images of 30 patients in treatment position and with the same field of view were used for the evaluation of the atlas and the conversion algorithms. The dose calculation accuracy of the generated sCTs was verified for volumetric modulated arc therapy (VMAT) and pencil beam scanning (PBS). VMAT and PBS plans were robust optimized on an internal target volume (ITV) against a patient set-up uncertainty of 5 mm. Average differences between CT and sCT dose calculations for the ITV V 95% were 0.5% (min 0.0%; max 5.0%) and 0.0% (min -0.1%; max 0.1%) for VMAT and PBS dose distributions, respectively. Average differences for the mean dose to the organs at risk were <0.2% (min -0.6%; max 1.2%) and <0.2% (min -2.0%; max 2.6%) for VMAT and PBS dose distributions, respectively. Results show that MRI-only photon and proton dose calculations are feasible for children with abdominal tumors.
Collapse
Affiliation(s)
- Filipa Guerreiro
- Department of Radiotherapy, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
194
|
Ouyang G, Liu J, Wang P, Ren Y, Yi P, Zhou Q, Chen J, Xiang B, Zhang Y, Zhang Z, Li L. Multiple factors affect the regeneration of liver. ACTA ACUST UNITED AC 2019; 64:791-798. [PMID: 30672999 DOI: 10.1590/1806-9282.64.09.791] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 01/20/2018] [Indexed: 11/21/2022]
Abstract
OBJECTIVE To study factors affecting the liver regeneration after hepatectomy. METHODS With 3D reconstitution technology, liver regeneration ability of 117 patients was analysed, and relative factors were studied. RESULTS There was no statistically difference between the volume of simulated liver resection and the actual liver resection. All livers had different degrees of regeneration after surgery. Age, gender and blood indicators had no impact on liver regeneration, while surgery time, intraoperative blood loss, blood flow blocking time and different ways of liver resection had a significant impact on liver regeneration; In addition, the patients' own pathological status, including, hepatitis and liver fibrosis all had a significant impact on liver regeneration. CONCLUSION 3D reconstitution model is a good model to calculate liver volume. Age, gender, blood indicators and biochemistry indicators have no impact on liver regeneration, but surgery indicators and patients' own pathological status have influence on liver regeneration.
Collapse
Affiliation(s)
- Gaoxiong Ouyang
- . Department of Hepatobiliary Surgery, Affiliated Cancer Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jianyong Liu
- . Department of Hepatobiliary Surgery, Affiliated Cancer Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Peng Wang
- . Department of Radiology, Affiliated Cancer Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yuan Ren
- . Department of Hepatobiliary Surgery, Affiliated Cancer Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Ping Yi
- . Department of Hepatobiliary Surgery, Affiliated Cancer Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Quan Zhou
- . Department of Hepatobiliary Surgery, Affiliated Cancer Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jun Chen
- . Department of Pathology, Affiliated Cancer Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Bangde Xiang
- . Department of Hepatobiliary Surgery, Affiliated Cancer Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yumei Zhang
- . Department of Hepatobiliary Surgery, Affiliated Cancer Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Zhiming Zhang
- . Department of Hepatobiliary Surgery, Affiliated Cancer Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Lequn Li
- . Department of Hepatobiliary Surgery, Affiliated Cancer Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
195
|
Abstract
PURPOSE OF REVIEW Our understanding of the biologic basis of neuroblastoma, the genetic heterogeneity of this malignancy and the role of host factors has expanded significantly in recent years. In this review, we highlight current and future risk-based treatment approaches and discuss the opportunities and challenges of selecting optimal therapies for specific patient subsets. RECENT FINDINGS Significant progress has been made in understanding neuroblastoma predisposition and new approaches have been taken to treatment of this disease. Although survival remains poor for patients with high-risk neuroblastoma, current-era therapy has improved outcomes. Integration of new prognostic markers into neuroblastoma classification systems will allow more precise risk classification and refined treatment assignment. Promising treatments that include targeted therapies as well as immunotherapeutics are being evaluated in clinical trials, and new predictive biomarkers are being developed. SUMMARY As our understanding of neuroblastoma biology deepens, our approaches to therapy for this disease continue to evolve. Improved risk stratification and the use of predictive biomarkers will aid in treatment selection for patients with neuroblastoma, and it is expected that future treatments will be associated with greater efficacy and less toxicity.
Collapse
|
196
|
Johnsen JI, Dyberg C, Wickström M. Neuroblastoma-A Neural Crest Derived Embryonal Malignancy. Front Mol Neurosci 2019; 12:9. [PMID: 30760980 PMCID: PMC6361784 DOI: 10.3389/fnmol.2019.00009] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/11/2019] [Indexed: 12/14/2022] Open
Abstract
Neuroblastoma is a neural crest derived malignancy of the peripheral nervous system and is the most common and deadliest tumor of infancy. It is characterized by clinical heterogeneity with a disease spectrum ranging from spontaneous regression without any medical intervention to treatment resistant tumors with metastatic spread and poor patient survival. The events that lead to the development of neuroblastoma from the neural crest have not been fully elucidated. Here we discuss factors and processes within the neural crest that when dysregulated have the potential to be initiators or drivers of neuroblastoma development. A more precise biological understanding of neuroblastoma causes and cell of origin is highly warranted. This will give valuable information for the development of medicines that specifically target molecules within neuroblastoma cells and also give hint about the mechanisms behind treatment resistance that is frequently seen in neuroblastoma.
Collapse
Affiliation(s)
- John Inge Johnsen
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet (KI), Stockholm, Sweden
| | - Cecilia Dyberg
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet (KI), Stockholm, Sweden
| | - Malin Wickström
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet (KI), Stockholm, Sweden
| |
Collapse
|
197
|
Larsson K, Kock A, Kogner P, Jakobsson PJ. Targeting the COX/mPGES-1/PGE 2 Pathway in Neuroblastoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1161:89-100. [PMID: 31562624 DOI: 10.1007/978-3-030-21735-8_9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The importance of prostaglandin E2 in cancer progression is well established, but research on its role in cancer has so far mostly been focused on epithelial cancer in adults while the knowledge about the contribution of prostaglandin E2 to childhood malignancies is limited. Neuroblastoma, an extracranial solid tumor of the sympathetic nervous system, mainly affects young children. Patients with tumors classified as high-risk have poor survival despite receiving intensive treatment, illustrating a need for new treatments complimenting existing ones. The basis of neuroblastoma treatment e.g. chemotherapy and radiation therapy, target the proliferating genetically unstable tumor cells leading to treatment resistance and relapses. The tumor microenvironment is an avenue, still to a great extent, unexplored and lacking effective targeted therapies. Cancer-associated fibroblasts is the main source of prostaglandin E2 in neuroblastoma contributing to angiogenesis, immunosuppression and tumor growth. Prostaglandin E2 is formed from its precursor arachidonic acid in a two-step enzymatic reaction. Arachidonic acid is first converted by cyclooxygenases into prostaglandin H2 and then further converted by microsomal prostaglandin E synthase-1 into prostaglandin E2. We believe targeting of microsomal prostaglandin E synthase-1 in cancer-associated fibroblasts will be an effective future therapeutic strategy in fighting neuroblastoma.
Collapse
Affiliation(s)
- Karin Larsson
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden.
| | - Anna Kock
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Per Kogner
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Per-Johan Jakobsson
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
198
|
Desai AV, Li Y, Getz K, Seif AE, Fisher B, Huang V, Aplenc R, Bagatell R. Resource utilization and toxicities after single versus tandem autologous stem cell rescue in high-risk neuroblastoma using a national administrative database. Pediatr Blood Cancer 2018; 65:e27372. [PMID: 30070014 DOI: 10.1002/pbc.27372] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 07/05/2018] [Accepted: 07/06/2018] [Indexed: 11/06/2022]
Abstract
BACKGROUND High-dose chemotherapy with autologous stem cell rescue (HDC-ASCR) has improved event-free survival for children with high-risk neuroblastoma. Common regimens include carboplatin/etoposide/melphalan (CEM), busulfan/melphalan (BuMel), and tandem HDC-ASCR [thiotepa/cyclophosphamide (TC) followed by CEM]. To complement clinical trial data and to evaluate the clinical burden associated with these regimens, resource ultilization (RU) was evaluated. An administrative database was used to evaluate RU in a previously developed high-risk neuroblastoma cohort. Single CEM and BuMel patients were followed for 60 days from the first day of the HDC-ASCR preparative regimen or until death, whichever came first. Tandem patients were followed from the first day of the first HDC-ASCR preparative regimen through day 60 from the first day of the second HDC-ASCR. Resources compared included inpatient days, ICU-level care, and medications administered. RESULTS A cohort of 578 patients was evaluated; 422 patients underwent single HDC-ASCR with CEM, 67 received BuMel, 72 underwent TC/CEM, and 17 received only the first portion of tandem HDC-ASCR. The median number of inpatient days and days of exposure to antibiotics, opioids, and total parenteral nutrition were higher in the tandem group than in the CEM and BuMel groups. However, the rate of use of several ICU-level resources per 1000 hospital days was lower for the tandem group. CONCLUSIONS These data suggest that while patients undergoing tandem HDC-ASCR were hospitalized longer, the severity of illness during hospitalization was not greater in tandem patients.
Collapse
Affiliation(s)
- Ami V Desai
- Pediatric Hematology/Oncology, University of Chicago Comprehensive Cancer Center, Chicago, Illinois, USA
| | - Yimei Li
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kelly Getz
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alix E Seif
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Brian Fisher
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Division of Infectious Diseases, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Center for Pediatric Clinical Effectiveness, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Vera Huang
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Richard Aplenc
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Center for Pediatric Clinical Effectiveness, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Rochelle Bagatell
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
199
|
Interleukin 2 with anti-GD2 antibody ch14.18/CHO (dinutuximab beta) in patients with high-risk neuroblastoma (HR-NBL1/SIOPEN): a multicentre, randomised, phase 3 trial. Lancet Oncol 2018; 19:1617-1629. [DOI: 10.1016/s1470-2045(18)30578-3] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/24/2018] [Accepted: 07/27/2018] [Indexed: 02/07/2023]
|
200
|
Arumugam S, Manning-Cork NJ, Gains JE, Boterberg T, Gaze MN. The Evidence for External Beam Radiotherapy in High-Risk Neuroblastoma of Childhood: A Systematic Review. Clin Oncol (R Coll Radiol) 2018; 31:182-190. [PMID: 30509728 DOI: 10.1016/j.clon.2018.11.031] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 10/27/2018] [Indexed: 12/15/2022]
Abstract
AIMS External beam radiotherapy is widely used in various ways in the management of neuroblastoma. Despite extensive clinical experience, the precise role of radiotherapy in neuroblastoma remains unclear. The purpose of this systematic review was to survey the published literature to identify, without bias, the evidence for the clinical effectiveness of external beam radiotherapy as part of the initial multimodality treatment of high-risk neuroblastoma. We considered four areas: treatment of the tumour bed and residual primary tumour, identification of any dose-response relationship, treatment of metastatic sites, identification of any technical advances that may be beneficial. We also aimed to define uncertainties, which may be clarified in future clinical trials. MATERIALS AND METHODS Bibliographic databases were searched for neuroblastoma and radiotherapy. Reviewers assessed 1283 papers for inclusion by title and abstract, with consensus achieved through discussion. Data extraction on 57 included papers was carried out by one reviewer and checked by another. Studies were assessed for their level of evidence and risk of bias, and a descriptive analysis of data was carried out. RESULTS Fifteen papers provided some evidence that radiotherapy to the tumour bed and residual tumour may possibly be of value. However, there is a significant risk of bias and no evidence that all subgroups will benefit. There is some suggestion from six papers that dose may be important, but no hard evidence. It remains unclear whether irradiation of metastatic sites is helpful. Technical advances may be of value in radiotherapy of high-risk neuroblastoma. CONCLUSIONS There are data that show that radiotherapy is of some efficacy in the management of high-risk neuroblastoma, but there is no level one evidence that shows that it is being used in the best possible way. Prospective randomised trials are necessary to provide more evidence to guide development of optimal radiotherapy treatment schedules.
Collapse
Affiliation(s)
- S Arumugam
- Department of Oncology, University College London Hospitals NHS Foundation Trust, London, UK
| | - N J Manning-Cork
- Department of Oncology, University College London Hospitals NHS Foundation Trust, London, UK
| | - J E Gains
- Department of Oncology, University College London Hospitals NHS Foundation Trust, London, UK
| | - T Boterberg
- Department of Radiation Oncology, Ghent University Hospital, Gent, Belgium
| | - M N Gaze
- Department of Oncology, University College London Hospitals NHS Foundation Trust, London, UK.
| |
Collapse
|